Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Curr Hypertens Rep ; 26(5): 213-224, 2024 May.
Article in English | MEDLINE | ID: mdl-38411777

ABSTRACT

PURPOSE OF REVIEW: The primary goal of this review article was to determine whether the three RAAS-associated SNPs, Renin-rs16853055, AGT-rs3789678 and ACE-rs4305 are genetically linked to the development of hypertension in preeclampsia. The secondary goal was to establish if there was a link between these SNPs and HIV infection. RECENT FINDINGS: There is a paucity of findings related to the aforementioned SNPs and preeclampsia. There are no recent findings on the rs16853055 renin polymorphism. The rs3789678 angiotensinogen polymorphism correlated significantly with gestational hypertension. The rs4305 ACE polymorphism showed no significant association with the development of pregnancy-induced hypertension. There are conflicting findings when determining the relationship between ethnicity and the predisposition of preeclampsia and hypertension in relation to the discussed RAAS-associated SNPs. To date, the association between RAAS-associated SNPs and preeclamptic women co-morbid with HIV in South Africa has revealed that certain alleles of the AGT gene are more prominent in HIV-infected PE compared to normotensive pregnant HIV-infected women.


Subject(s)
Angiotensinogen , HIV Infections , Peptidyl-Dipeptidase A , Polymorphism, Single Nucleotide , Pre-Eclampsia , Renin-Angiotensin System , Renin , Humans , Pregnancy , Female , Pre-Eclampsia/genetics , HIV Infections/genetics , HIV Infections/complications , Polymorphism, Single Nucleotide/genetics , Angiotensinogen/genetics , Renin-Angiotensin System/genetics , Renin/genetics , Peptidyl-Dipeptidase A/genetics , Genetic Predisposition to Disease , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/virology
2.
Bull Exp Biol Med ; 175(2): 260-264, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37474692

ABSTRACT

We studied the relative length of telomeres in newborns with unrealized perinatal transmission of HIV (zero viral load according to PCR results). A cross-sectional survey of 62 newborns of HIV-infected mothers (Apgar score 8); the control group consisted of 80 healthy newborns (Apgar score 8). DNA extracted from whole venous blood samples was analyzed. In newborns of HIV-infected mothers, the relative length of telomeres was significantly lower (0.69 (0.66; 0.72)) than in newborns of the control group (1.1 (0.97; 1.22)) (p<0.001). No significant differences in the relative length of telomeres were found between newborns of mothers with a viral load at the time of delivery and with undetectable viral load: 0.69 (0.66; 0.73) and 0.69 (0.63; 0.72). These findings indicate that HIV-infection in mothers or exposure to antiretroviral therapy has an impact on the relative telomere length in leukocytes of newborns.


Subject(s)
HIV Infections , Pregnancy Complications, Infectious , Pregnancy , Female , Infant, Newborn , Humans , Mothers , Infectious Disease Transmission, Vertical/prevention & control , Cross-Sectional Studies , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/prevention & control , Leukocytes , Telomere/genetics , Pregnancy Complications, Infectious/genetics
3.
Nat Cell Biol ; 25(7): 1047-1060, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37400500

ABSTRACT

During pregnancy the maternal-fetal interface plays vital roles in fetal development. Its disruption is frequently found in pregnancy complications. Recent studies show increased incidences of adverse pregnancy outcomes in patients with COVID-19; however, the mechanism remains unclear. Here we analysed the molecular impacts of SARS-CoV-2 infection on the maternal-fetal interface. Generating bulk and single-nucleus transcriptomic and epigenomic profiles from patients with COVID-19 and control samples, we discovered aberrant immune activation and angiogenesis patterns in distinct cells from patients. Surprisingly, retrotransposons were also dysregulated in specific cell types. Notably, reduced enhancer activities of LTR8B elements were functionally linked to the downregulation of pregnancy-specific glycoprotein genes in syncytiotrophoblasts. Our findings revealed that SARS-CoV-2 infection induced substantial changes to the epigenome and transcriptome at the maternal-fetal interface, which may be associated with pregnancy complications.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Pregnancy , Female , Humans , COVID-19/genetics , Transcriptome , SARS-CoV-2 , Epigenomics , Pregnancy Complications, Infectious/genetics , Single-Cell Analysis
4.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166768, 2023 10.
Article in English | MEDLINE | ID: mdl-37269984

ABSTRACT

A unique immunological condition, pregnancy ensures fetus from maternal rejection, allows adequate fetal development, and protects against microorganisms. Infections during pregnancy may lead to devastating consequences for pregnant women and fetuses, resulting in the mother's death, miscarriage, premature childbirth, or neonate with congenital infection and severe diseases and defects. Epigenetic (heritable changes in gene expression) mechanisms like DNA methylation, chromatin modification, and gene expression modulation during gestation are linked with the number of defects in the fetus and adolescents. The feto-maternal crosstalk for fetal survival during the entire gestational stages are tightly regulated by various cellular pathways, including epigenetic mechanisms that respond to both internal as well outer environmental factors, which can influence the fetal development across the gestational stages. Due to the intense physiological, endocrinological, and immunological changes, pregnant women are more susceptible to bacterial, viral, parasitic, and fungal infections than the general population. Microbial infections with viruses (LCMV, SARS-CoV, MERS-CoV, and SARS-CoV-2) and bacteria (Clostridium perfringens, Coxiella burnetii, Listeria monocytogenes, Salmonella enteritidis) further increase the risk to maternal and fetal life and developmental outcome. If the infections remain untreated, the possibility of maternal and fetal death exists. This article focused on the severity and susceptibility to infections caused by Salmonella, Listeria, LCMV, and SARS-CoV-2 during pregnancy and their impact on maternal health and the fetus. How epigenetic regulation during pregnancy plays a vital role in deciding the fetus's developmental outcome under various conditions, including infection and other stress. A better understanding of the host-pathogen interaction, the characterization of the maternal immune system, and the epigenetic regulations during pregnancy may help protect the mother and fetus from infection-mediated outcomes.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Infant, Newborn , Adolescent , Pregnancy , Female , Humans , Pregnancy Complications, Infectious/genetics , COVID-19/genetics , SARS-CoV-2 , Epigenesis, Genetic , Fetal Development
5.
J Cell Mol Med ; 27(11): 1465-1476, 2023 06.
Article in English | MEDLINE | ID: mdl-37078407

ABSTRACT

There is a growing body of evidence that innate immunity also plays an important role in the progression of hepatitis B virus (HBV) infection. However, there is less study on systematically elucidating the characteristics of innate immunity in HBV-infected pregnant women. We compared the features of peripheral blood mononuclear cells in three healthy pregnant women and three HBV-infected pregnant women by single-cell RNA sequencing. 10 DEGs were detected between groups and monocytes were the main expression source of most of the DEGs, which involved in the inflammatory response, apoptosis and immune regulation. Meanwhile, qPCR and ELISA were performed to verify above genes. Monocytes displayed immune response defect, reflecting poor ability of response to IFN. In addition, eight clusters were identified in monocytes. We identified molecular drivers in monocytes subpopulations.TNFSF10+ monocytes, MT1G+ monocytes and TUBB1+ monocytes were featured with different gene expression pattern and biological function.TNFSF10+ monocytes and MT1G+ monocytes were characterized by high levels of inflammation response.TNFSF10+ monocytes, MT1G+ monocytes and TUBB1+ monocytes showed decreased response to IFN. Our results dissects alterations in monocytes related to the immune response of HBV-infected pregnant women and provides a rich resource for fully understanding immunopathogenesis and developing effective preventing HBV intrauterine infection strategies.


Subject(s)
Hepatitis B , Pregnancy Complications, Infectious , Humans , Pregnancy , Female , Hepatitis B virus/genetics , Monocytes , Pregnant Women , Leukocytes, Mononuclear/metabolism , Hepatitis B Surface Antigens , Pregnancy Complications, Infectious/genetics , Hepatitis B/genetics , Hepatitis B/metabolism , Sequence Analysis, RNA
6.
Ann Diagn Pathol ; 62: 152080, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36535188

ABSTRACT

Novel biomarkers of in utero infections are needed to help guide early therapy. The toll like receptors (TLRs) and retinoic acid-inducible gene 1 (RIG-1) are proteins involved in the initial reaction of the innate immune system to infectious diseases. This study tested the hypothesis that a panel of TLRs and RIG-1 in the placenta could serve as an early biomarker of in utero infections. The TLRs and RIG-1 expression as determined by immunohistochemistry was scored in 10 control placentas (normal delivery or neonatal damage from known non-infectious cause), 8 placentas from documented in utero bacterial infection, and 7 placentas from documented in utero viral infections blinded to the clinical information. The non-infected placentas showed the following profile: no expression (TLR1, TLR3, TLR4, TLR7, TLR8), moderate expression (TLR2), and strong expression (RIG-1). The bacterial and viral infection cases shared the following profile: no to mild expression (TLR 2, TLR7, and RIG1), moderate expression (TLR4), and strong expression (TLR1, TLR3, and TLR8). The histologic findings in the chorionic villi were equivalent in the infected cases and controls, underscoring the need for molecular testing by the surgical pathologist when in utero infection is suspected. The results suggest that a panel of TLRs/RIG-1 analyses can allow the pathologist and/or clinician to diagnose in utero infections soon after birth. Also, treatments to antagonize the effects of TLR1, 3, and 8 may help abrogate in utero neonatal damage.


Subject(s)
Placenta , Pregnancy Complications, Infectious , Female , Humans , Infant, Newborn , Pregnancy/immunology , Placenta/immunology , Placenta/metabolism , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4 , Toll-Like Receptor 7 , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/metabolism
7.
Am J Obstet Gynecol ; 227(4): 634.e1-634.e12, 2022 10.
Article in English | MEDLINE | ID: mdl-35609640

ABSTRACT

BACKGROUND: Congenital cytomegalovirus infection is the most common perinatal infection and a significant cause of sensorineural hearing loss, cerebral palsy, and neurodevelopmental disability. There is a paucity of human gene expression studies examining the pathophysiology of cytomegalovirus infection. OBJECTIVE: This study aimed to perform a whole transcriptomic assessment of amniotic fluid from pregnancies with live fetuses to identify differentially expressed genes and enriched Gene Ontology categories associated with congenital cytomegalovirus infection. STUDY DESIGN: Amniotic fluid supernatant was prospectively collected from pregnant women undergoing amniocentesis for suspected congenital cytomegalovirus infection because of first-trimester maternal primary infection or ultrasound features suggestive of fetal infection. Women who had received therapy to prevent fetal infection were excluded. Congenital cytomegalovirus infection was diagnosed via viral polymerase chain reaction of amniotic fluid; cytomegalovirus-infected fetuses were paired with noninfected controls, matched for gestational age and fetal sex. Paired-end RNA sequencing was performed on amniotic fluid cell-free RNA with the Novaseq 6000 at a depth of 30 million reads per sample. Following quality control and filtering, reads were mapped to the human genome and counts summarized across genes. Differentially expressed genes were identified using 2 approaches: voomWithQualityWeights in conjunction with limma and RUVSeq with edgeR. Genes with a false discovery rate <0.05 were considered statistically significant. Differential exon use was analyzed using DEXSeq. Functional analysis was performed using gene set enrichment analysis and Ingenuity Pathway Analysis. Manual curation of differentially regulated genes was also performed. RESULTS: Amniotic fluid samples were collected from 50 women; 16 (32%) had congenital cytomegalovirus infection confirmed by polymerase chain reaction. After excluding 3 samples without matched controls, 13 cytomegalovirus-infected samples collected at 18 to 23 weeks and 13 cytomegalovirus-negative gestation-matched controls were submitted for RNA sequencing and analysis (N=26). Ten of the 13 pregnancies with cytomegalovirus-infected fetuses had amniocentesis because of serologic evidence of maternal primary infection with normal fetal ultrasound, and 3 had amniocentesis because of ultrasound abnormality suggestive of cytomegalovirus infection. Four cytomegalovirus-infected pregnancies ended in termination (n=3) or fetal death (n=1), and 9 resulted in live births. Pregnancy outcomes were available for 11 of the 13 cytomegalovirus-negative controls; all resulted in live births of clinically-well infants. Differential gene expression analysis revealed 309 up-regulated and 32 down-regulated genes in the cytomegalovirus-infected group compared with the cytomegalovirus-negative group. Gene set enrichment analysis showed significant enrichment of multiple Gene Ontology categories involving the innate immune response to viral infection and interferon signaling. Of the 32 significantly down-regulated genes, 8 were known to be involved in neurodevelopment and preferentially expressed by the brain. Six specific cellular restriction factors involved in host defense to cytomegalovirus infection were up-regulated in the cytomegalovirus-infected group. Ingenuity Pathway Analysis predicted the activation of pathways involved in progressive neurologic disease and inflammatory neurologic disease. CONCLUSION: In this next-generation sequencing study, we revealed new insights into the pathophysiology of congenital cytomegalovirus infection. These data on the up-regulation of the intraamniotic innate immune response to cytomegalovirus infection and the dysregulation of neurodevelopmental genes may inform future approaches to developing prognostic markers and assessing fetal responses to in utero therapy.


Subject(s)
Cell-Free Nucleic Acids , Cytomegalovirus Infections , Pregnancy Complications, Infectious , Amniotic Fluid/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/genetics , Female , Humans , Infant , Interferons/genetics , Interferons/metabolism , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/metabolism , RNA-Seq
8.
Exp Cell Res ; 414(2): 113086, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35283101

ABSTRACT

In 2015, Brazil reported an outbreak identified as Zika virus (ZIKV) infection associated with congenital abnormalities. To date, a total of 86 countries and territories have described evidence of Zika infection and recently the appearance of the African ZIKV lineage in Brazil highlights the risk of a new epidemic. The spectrum of ZIKV infection-induced alterations at both cellular and molecular levels is not completely elucidated. Here, we present for the first time the gene expression responses associated with prenatal ZIKV infection from ocular cells. We applied a recently developed non-invasive method (impression cytology) which use eye cells as a model for ZIKV studies. The ocular profiling revealed significant differences between exposed and control groups, as well as a different pattern in ocular transcripts from Congenital Zika Syndrome (CZS) compared to ZIKV-exposed but asymptomatic infants. Our data showed pathways related to mismatch repair, cancer, and PI3K/AKT/mTOR signaling and genes probably causative or protective in the modulation of ZIKV infection. Ocular cells revealed the effects of ZIKV infection on primordial neuronal cell genes, evidenced by changes in genes associated with embryonic cells. The changes in gene expression support an association with the gestational period of the infection and provide evidence for the resulting clinical and ophthalmological pathologies. Additionally, the findings of cell death- and cancer-associated deregulated genes raise concerns about the early onset of other potential pathologies including the need for tumor surveillance. Our results thus provide direct evidence that infants exposed prenatally to the Zika virus, not only with CZS but also without clinical signs (asymptomatic) express cellular and molecular changes with potential clinical implications.


Subject(s)
Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus , Eye/pathology , Female , Humans , Infant , Phosphatidylinositol 3-Kinases , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/genetics , Zika Virus/genetics , Zika Virus Infection/diagnosis , Zika Virus Infection/epidemiology , Zika Virus Infection/genetics
9.
Afr Health Sci ; 22(4): 428-434, 2022 Dec.
Article in English | MEDLINE | ID: mdl-37092059

ABSTRACT

Background: Pre-treatment HIV drug resistance is a threat to elimination of mother to child HIV transmission and could lead to virological failure among HIV-positive pregnant women. We analysed genotypic HIV drug resistance (HIVDR) of baseline samples of participants enrolled in the Option B+ clinical trial in Uganda. Methods: HIV-infected pregnant women attending antenatal care were enrolled from Uganda's National Referral Hospital (Mulago) and Mityana District general hospital and surrounding health centers (HCs). Genotypic HIV testing was performed on blood samples from the first 135 enrolled women out of a subset of 136 participants (25%) who had a baseline VL>1000 copies/mL as one sample failed to amplify. Results: 159/540 (29.4%) had a VL < 1000 copies/ml and 381/540 (70.6%) had a VL >1,000 copies/ml. Of the women with VL>1000 copies/ml, 32 (23.7%) had resistance mutations including 29/135 (21.5%) NNRTI mutations, 6/135 (4.4%) NRTI mutations and 3/135 (2.2%) had both NNRTI and NRTI mutations. The most common NNRTI resistance mutations were: K103KN (5), K103N (5), V179T (4) and E138A (4). Conclusions: One quarter of the HIV-infected pregnant women in this trial at baseline had NNRTI genotypic resistance mutations. Our findings support new WHO guidelines for first-line ART that were changed to dolutegravir-based regimens.


Subject(s)
Anti-HIV Agents , Drug Resistance, Viral , HIV Infections , HIV-1 , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious , Female , Humans , Pregnancy , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Drug Resistance, Viral/genetics , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/transmission , HIV-1/genetics , Infectious Disease Transmission, Vertical/prevention & control , Mutation , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/prevention & control , Prenatal Care , Uganda , Viral Load/genetics
10.
Front Immunol ; 12: 764746, 2021.
Article in English | MEDLINE | ID: mdl-34899713

ABSTRACT

Host factors that influence Congenital Zika Syndrome (CZS) outcome remain elusive. Interferons have been reported as the main antiviral factor in Zika and other flavivirus infections. Here, we accessed samples from 153 pregnant women (77 without and 76 with CZS) and 143 newborns (77 without and 66 with CZS) exposed to ZIKV conducted a case-control study to verify whether interferon alfa receptor 1 (IFNAR1) and interferon lambda 2 and 4 (IFNL2/4) single nucleotide polymorphisms (SNPs) contribute to CZS outcome, and characterized placenta gene expression profile at term. Newborns carrying CG/CC genotypes of rs2257167 in IFNAR1 presented higher risk of developing CZS (OR=3.41; IC=1.35-8.60; Pcorrected=0.032). No association between IFNL SNPs and CZS was observed. Placenta from CZS cases displayed lower levels of IFNL2 and ISG15 along with higher IFIT5. The rs2257167 CG/CC placentas also demonstrated high levels of IFIT5 and inflammation-related genes. We found CZS to be related with exacerbated type I IFN and insufficient type III IFN in placenta at term, forming an unbalanced response modulated by the IFNAR1 rs2257167 genotype. Despite of the low sample size se findings shed light on the host-pathogen interaction focusing on the genetically regulated type I/type III IFN axis that could lead to better management of Zika and other TORCH (Toxoplasma, Others, Rubella, Cytomegalovirus, Herpes) congenital infections.


Subject(s)
Interleukins/immunology , Pregnancy Complications, Infectious/immunology , Receptor, Interferon alpha-beta/immunology , Zika Virus Infection/immunology , Female , Genotype , Humans , Infant, Newborn , Interleukins/genetics , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology , Pregnancy , Pregnancy Complications, Infectious/genetics , Receptor, Interferon alpha-beta/genetics , Zika Virus Infection/genetics
11.
Viruses ; 13(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34835060

ABSTRACT

Congenital Zika syndrome (CZS) is characterized by a diverse group of congenital malformations induced by ZIKV infection during pregnancy. Type III interferons have been associated with placental immunity against ZIKV and restriction of vertical transmission in mice, and non-coding single-nucleotide polymorphisms (SNPs) on these genes are well known to influence susceptibility to other viral infections. However, their effect on ZIKV pathogenesis has not yet been explored. To investigate whether maternal non-coding SNPs at IFNL genes are associated with CZS, 52 women infected with ZIKV during pregnancy were enrolled in a case-control association study. A total of 28 women were classified as cases and 24 as controls based on the presence or absence of CZS in their infants, and seven Interferon-λ non-coding SNPs (rs12980275, rs8099917, rs4803217, rs4803219, rs8119886, rs368234815, rs12979860) were genotyped. The results of logistic regression analyses show an association between the G allele at rs8099917 and increased susceptibility to CZS under a log-additive model (adjustedOR = 2.80; 95%CI = 1.14-6.91; p = 0.02), after adjustment for trimester of infection and genetic ancestry. These results provide evidence of an association between Interferon-λ SNPs and CZS, suggesting rs8099917 as a promising candidate for further studies on larger cohorts.


Subject(s)
Interferons/genetics , Pregnancy Complications, Infectious/genetics , Zika Virus Infection/congenital , Zika Virus Infection/genetics , Alleles , Brazil , Case-Control Studies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Polymorphism, Single Nucleotide , Pregnancy , Zika Virus
12.
Biomolecules ; 11(8)2021 08 03.
Article in English | MEDLINE | ID: mdl-34439812

ABSTRACT

Intra-amniotic infections (IAI) are one of the reasons for preterm birth. High mobility group box 1 (HMGB1) is a nuclear protein with various physiological functions, including tissue healing. Its excessive extracellular release potentiates inflammatory reaction and can revert its action from beneficial to detrimental. We infected the amniotic fluid of a pig on the 80th day of gestation with 1 × 104 colony forming units (CFUs) of E. coli O55 for 10 h, and evaluated the appearance of HMGB1, receptor for glycation endproducts (RAGE), and Toll-like receptor (TLR) 4 in the amniotic membrane and fluid. Sham-infected amniotic fluid served as a control. The expression and release of HMGB1 were evaluated by Real-Time PCR, immunofluorescence, immunohistochemistry, and ELISA. The infection downregulated HMGB1 mRNA expression in the amniotic membrane, changed the distribution of HMGB1 protein in the amniotic membrane, and increased its level in amniotic fluid. All RAGE mRNA, protein expression in the amniotic membrane, and soluble RAGE level in the amniotic fluid were downregulated. TLR4 mRNA and protein expression and soluble TLR4 were all upregulated. HMGB1 is a potential target for therapy to suppress the exaggerated inflammatory response. This controlled expression and release can, in some cases, prevent the preterm birth of vulnerable infants. Studies on suitable animal models can contribute to the development of appropriate therapy.


Subject(s)
Escherichia coli Infections/veterinary , Escherichia coli/pathogenicity , HMGB1 Protein/genetics , Pregnancy Complications, Infectious/veterinary , RNA, Messenger/genetics , Receptor for Advanced Glycation End Products/genetics , Toll-Like Receptor 4/genetics , Amnion/immunology , Amnion/microbiology , Amnion/pathology , Amniotic Fluid/immunology , Amniotic Fluid/microbiology , Animals , Disease Models, Animal , Escherichia coli/growth & development , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Gene Expression Regulation , HMGB1 Protein/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Pregnancy , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/microbiology , Premature Birth/prevention & control , RNA, Messenger/immunology , Receptor for Advanced Glycation End Products/immunology , Signal Transduction , Swine , Toll-Like Receptor 4/immunology
13.
Cells ; 10(7)2021 07 08.
Article in English | MEDLINE | ID: mdl-34359894

ABSTRACT

COVID-19 is associated with increased incidence of preterm birth (PTB). We assessed pathways by which SARS-CoV-2 could access the placenta. Placentae, from PTB with or without chorioamnionitis (ChA), or from term pregnancies (n = 12/13/group) were collected. Peripheral blood was collected from healthy pregnant women (n = 6). Second trimester placental explants (16-20 weeks, n = 5/group) were treated with lipopolysaccharide (LPS, to mimic bacterial infection) and ACE2, CCL2, IL-6/8 and TNFα mRNA was assessed. ChA-placentae exhibited increased ACE2 and CCL2 mRNA expression (p < 0.05). LPS increased cytokine and ACE2 mRNA in placental explants. Placental ACE2 protein localized to syncytiotrophoblast, fetal endothelium, extravillous trophoblast and in immune cells-subsets (M1/M2 macrophage and neutrophils) within the villous stroma. Significantly increased numbers of M1 macrophage and neutrophils were present in the ChA-placenta (p < 0.001). Subsets of peripheral immune cells from pregnant women express the ACE2 mRNA and protein. A greater fraction of granulocytes was positive for ACE2 protein expression compared to lymphocytes or monocytes. These data suggest that in pregnancies complicated by ChA, ACE2 positive immune cells in the maternal circulation have the potential to traffic SARS-CoV-2 virus to the placenta and increase the risk of vertical transmission to the placenta/fetus.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Gene Expression , Placenta/metabolism , Pregnancy Complications, Infectious/genetics , Premature Birth/etiology , Adult , COVID-19/genetics , COVID-19/transmission , Female , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Lymphocytes/metabolism , Monocytes/metabolism , Placenta/cytology , Pregnancy , Premature Birth/genetics , SARS-CoV-2/isolation & purification
14.
Cells ; 10(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34359957

ABSTRACT

MicroRNAs are gene expression regulators associated with several human pathologies, including those generated by viral infections. Their role in SARS-CoV-2 infection and COVID-19 has been investigated and reviewed in many informative studies; however, a thorough miRNA outline in SARS-CoV-2-infected pregnant women (SIPW), at both systemic and placental levels, is missing. To fill this gap, blood and placenta biopsies collected at delivery from 15 asymptomatic SIPW were immediately analysed for: miRNA expression (n = 84) (QPCR array), antiviral/immune mRNA target expression (n = 74) (QGene) and cytokine/chemokines production (n = 27) (Multiplex ELISA). By comparing these results with those obtained from six uninfected pregnant women (UPW), we observed that, following SARS-CoV-2 infection, the transcriptomic profile of pregnant women is significantly altered in different anatomical districts, even in the absence of clinical symptoms and vertical transmission. This characteristic combination of miRNA and antiviral/immune factors seems to control both the infection and the dysfunctional immune reaction, thus representing a positive correlate of protection and a potential therapeutic target against SARS-CoV-2.


Subject(s)
COVID-19/genetics , MicroRNAs/genetics , Pregnancy Complications, Infectious/genetics , Adult , COVID-19/blood , COVID-19/diagnosis , Female , Humans , MicroRNAs/analysis , MicroRNAs/blood , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/diagnosis , SARS-CoV-2/isolation & purification , Transcriptome , Young Adult
15.
PLoS One ; 16(7): e0255054, 2021.
Article in English | MEDLINE | ID: mdl-34293039

ABSTRACT

BACKGROUND: Hepatitis E virus (HEV) infection is endemic in Bangladesh and there are occasional outbreaks. The molecular characteristics and pathogenesis of endemic and outbreak HEV strains are poorly understood. We compared the genetic relatedness and virulence associated mutations of endemic HEV strains with outbreak strains. METHODS: We analyzed systematically collected serum samples from HEV immunoglobulin M (IgM) positive patients attended at Bangabandhu Sheikh Mujib Medical University, Dhaka from August 2013 to June 2015. HEV RNA positive samples were subjected to whole genome sequencing. Genotype and subtype of the strains were determined by phylogenetic analysis. Virulence associated mutations e.g. acute viral hepatitis (AVH), fulminant hepatic failure (FHF), chronic hepatitis, ribavirin treatment failure (RTF), B and T cell neutralization epitopes were determined. RESULTS: 92 HEV immunoglobulin M (IgM) antibody positive plasma samples (43 in 2013-2014 and 49 in 2014-2015) were studied. 77.1% (70/92) of the samples were HEV RNA positive. A 279 bp open reading frame (ORF) 2 and ORF 3 sequence was obtained from 54.2% (38/70) of the strains. Of these 38 strains, whole genome sequence (WGS) was obtained from 21 strains. In phylogenetic analysis of 38 (279 bp) sequence all HEV sequences belonged to genotype 1 and subtype 1a. Further phylogenetic analysis of 21 HEV WGS, Bangladeshi HEV sequences clustered with genotype 1a sequences from neighboring countries. Within genotype 1a cluster, Bangladesh HEV strains formed a separate cluster with the 2010 HEV outbreak strains from northern Bangladesh. 80.9 to 100% of the strains had A317T, T735I, L1120I, L1110F, P259S, V1479I, G1634K mutations associates AVH, FHF and RTF. Mutations in T cell recognition epitope T3, T5, T7 was observed in 76.1%, 100% and 100% of the strains respectively. CONCLUSION: Strains of HEV genotype 1a are dominant in Bangladesh and are associated with endemic and outbreak of HEV infection. HEV isolates in Bangladesh have high prevalence of virulence associated mutations and mutation which alters antigenicity to B and T cell epitopes.


Subject(s)
Disease Outbreaks , Endemic Diseases , Genotype , Hepatitis E virus , Hepatitis E , Phylogeny , Pregnancy Complications, Infectious , Adult , Bangladesh/epidemiology , Cross-Sectional Studies , Female , Hepatitis Antibodies/blood , Hepatitis E/blood , Hepatitis E/epidemiology , Hepatitis E/genetics , Hepatitis E virus/genetics , Hepatitis E virus/metabolism , Humans , Immunoglobulin M/blood , Liver Failure, Acute/blood , Liver Failure, Acute/epidemiology , Liver Failure, Acute/genetics , Male , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/genetics , Prospective Studies
16.
Fertil Steril ; 116(4): 1030-1039, 2021 10.
Article in English | MEDLINE | ID: mdl-34325918

ABSTRACT

OBJECTIVE: To study the inflammatory profile and genes involved in the response to bacterial infections in women who developed spontaneous abortion in the presence of Ureaplasma parvum. DESIGN: Cross-sectional study. SETTING: A maternal and child referral center. PATIENT(S): Eighty-nine women with spontaneous abortion and 20 women with normal vaginal delivery (control group) were studied. INTERVENTION(S): Samples of biopsied placental tissue were collected for Mollicutes detection. MAIN OUTCOME MEASURE(S): The samples were subjected to histologic analysis, immunohistochemical evaluation for macrophages and lymphocytes, cytokine quantification, and quantitative polymerase chain reaction array to evaluate the expression of 84 genes related to the innate and adaptive immune responses. RESULT(S): The presence of U. parvum in the abortion group was positively associated with the influx of polymorphonuclear cells in the placental tissue and increased concentrations of interleukin-6 and interleukin-12p70. U. parvum caused downregulation of genes involved in the immune response, such as attraction of immune cells, activation of an inflammatory response, T-helper cell 17 response activation, and activation of the complement system at the beginning and end of pregnancy. CONCLUSION: The direct action of U. parvum on placental tissue altered the gestational tolerogenic state, reducing the immune response against pathogens and activating the extrinsic apoptotic pathway, causing spontaneous abortion.


Subject(s)
Abortion, Spontaneous/microbiology , Histocompatibility, Maternal-Fetal , Immune Tolerance , Placenta/microbiology , Pregnancy Complications, Infectious/microbiology , Ureaplasma Infections/microbiology , Ureaplasma/pathogenicity , Abortion, Spontaneous/diagnosis , Abortion, Spontaneous/immunology , Adaptive Immunity , Apoptosis , Apoptosis Regulatory Proteins/genetics , Case-Control Studies , Cross-Sectional Studies , Cytokines/genetics , Female , Gene Expression Regulation , Histocompatibility, Maternal-Fetal/genetics , Host-Pathogen Interactions , Humans , Immune Tolerance/genetics , Immunity, Innate , Placenta/immunology , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/immunology , Risk Factors , Ureaplasma/immunology , Ureaplasma Infections/diagnosis , Ureaplasma Infections/genetics , Ureaplasma Infections/immunology
17.
PLoS One ; 16(6): e0252309, 2021.
Article in English | MEDLINE | ID: mdl-34170911

ABSTRACT

INTRODUCTION: Maternal Cytomegalovirus (CMV) infection in the first trimester (T1) of pregnancy is a public health concern, as it increases the risk of severe neurodevelopmental outcomes associated with congenital infection compared to infections occurring later during pregnancy. OBJECTIVES: To determine CMV seroprevalence in T1 of pregnancy, its trend, risk factors and the incidence rate of primary infection during pregnancy. METHODS: Using the biobank of the prospective cohort "Grossesse en Santé de Québec" collected between April 2005 and March 2010 at the Québec-Laval Hospital, Québec, Canada, maternal CMV serology was determined using Abbott Architect Chemiluminescence microparticle immunoassays for immunoglobulin G(IgG), immunoglobulin M(IgM) titration and IgG avidity testing. Changepoint detection analysis was used to assess temporal trends. Risk factors associated with seropositivity were determined by multivariable logistic regression. RESULTS: CMV seroprevalence in T1 of pregnancy was 23.4% (965/4111, 95% CI, 22.1-24.7%). The incidence rate for CMV primary infection during pregnancy was 1.8 (95% CI, 1.2-2.6) per 100 person-years. No changepoint was identified in the maternal CMV-seroprevalence trend. Multivariable analyses showed that T1 maternal CMV seropositivity was associated with having one child OR 1.3 (95% CI, 1.10-1.73) or two or more children OR 1.5 (95%CI, 1.1-2.1), ethnicity other than Caucasian OR 2.1 (95% CI, 1.1-3.8) and country of birth other than Canada and the USA OR 2.8 (95% CI, 1.5-4.9). CONCLUSIONS: In this cohort, maternal seroprevalence in T1 of pregnancy and seroconversion rate were low. This information and identified risk factors could help guide the development and implementation of preventive actions and evidence-based health policies to prevent CMV infection during pregnancy.


Subject(s)
Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/virology , Pregnancy Complications, Infectious/genetics , Adolescent , Adult , Antibodies, Viral/immunology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Female , Fetal Diseases/etiology , Fetal Diseases/immunology , Fetal Diseases/virology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Infectious Disease Transmission, Vertical , Male , Parturition/immunology , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Pregnancy Trimester, First/immunology , Prospective Studies , Quebec , Risk Factors , Seroepidemiologic Studies , Young Adult
18.
PLoS Negl Trop Dis ; 15(6): e0009507, 2021 06.
Article in English | MEDLINE | ID: mdl-34125832

ABSTRACT

Congenital Zika Syndrome (CZS) is a critical illness with a wide range of severity caused by Zika virus (ZIKV) infection during pregnancy. Life-threatening neurodevelopmental dysfunctions are among the most common phenotypes observed in affected newborns. Risk factors that contribute to susceptibility and response to ZIKV infection may be related to the virus itself, the environment, and maternal genetic background. Nevertheless, the newborn's genetic contribution to the critical illness is still not elucidated. Here, we aimed to identify possible genetic variants as well as relevant biological pathways that might be associated with CZS phenotypes. For this purpose, we performed a whole-exome sequencing in 40 children born to women with confirmed exposure to ZIKV during pregnancy. We investigated the occurrence of rare harmful single-nucleotide variants (SNVs) possibly associated with inborn errors in genes ontologically related to CZS phenotypes. Moreover, an exome-wide association analysis was also performed using a case-control design (29 CZS cases and 11 controls), for both common and rare variants. Five out of the 29 CZS patients harbored known pathogenic variants likely to contribute to mild to severe manifestations observed. Approximately, 30% of affected individuals carried at least one pathogenic or likely pathogenic SNV in genes candidates to play a role in CZS. Our common variant association analysis detected a suggestive protective effect of the rs2076469 in DISP3 gene (p-value: 1.39 x 10-5). The IL12RB2 gene (p-value: 2.18x10-11) also showed an unusual distribution of nonsynonymous rare SNVs in control samples. Finally, genes harboring harmful variants are involved in processes related to CZS phenotypes such as neurological development and immunity. Therefore, both rare and common variations may be likely to contribute as the underlying genetic cause of CZS susceptibility. The variations and pathways identified in this study may also have implications for the development of therapeutic strategies in the future.


Subject(s)
Genetic Predisposition to Disease , Pregnancy Complications, Infectious/virology , Zika Virus Infection/congenital , Zika Virus Infection/genetics , Brazil , Case-Control Studies , Female , Humans , Infant, Newborn , Male , Polymorphism, Single Nucleotide , Pregnancy , Pregnancy Complications, Infectious/genetics , Exome Sequencing , Zika Virus/physiology
19.
J Reprod Immunol ; 146: 103342, 2021 08.
Article in English | MEDLINE | ID: mdl-34102513

ABSTRACT

This study evaluated the impact of the TLR7 Gln11Leu (rs179008) and TLR9 -1237 T/C (rs5743836) single nucleotide polymorphisms (SNPs) on susceptibility to placental infections and pregnancy complications in 455 Brazilian women. Demographic, socioeconomic, gynecological, and clinical characteristics of the women were collected. Placental tissues were sampled from pregnant women and human and viral DNA was extracted. Human alphaherpesvirus 1 (Herpes simplex virus type 1, HSV-1), Human alphaherpesvirus 2 (Herpes simplex virus type 2, HSV-2) and Human betaherpesvirus 5 (Human cytomegalovirus, HCMV) were detected by nested PCR. TLR9 and TLR7 SNPs were genotyped by PCR amplification of bi-directional specific alleles (Bi-PASA) and restriction fragment length polymorphism (RFLP), respectively. Infections at the time of birth were detected in 45.71 % of women. The presence of the TT genotype (recessive model) of the TLR7 SNP was associated with increased susceptibility to HSV-1 infection (O.R. = 2.23, p = 0.05). The presence of the C allele of the TLR9 SNP, in heterozygosis or homozygosis (dominant model), decreased the infection risk by HCMV (O.R. = 0.31, p-mod<0.05). The TT genotype (recessive model) of the TLR7 SNP was significantly associated (p < 0.05) with increased occurrence of pre-treated hypertension. The codominant model of the TLR9 SNP was significantly associated (p < 0.05) with reduced risk of hospitalization during pregnancy. In combination, the AA/CT (TLR7-TLR9) genotypes significantly decreased the risk of placental infection by HSV-1 and/or HSV-2 (O.R. = 0.47, p = 0.02), the susceptibility to all infectious agents considered in combination (O.R. = 0.4, p = 0.00), and the need of hospitalization (O.R. = 0.48, p = 0.02). In conclusion, TLR7 and TLR9 SNPs are potential modulating factors for the risk of placental infections and pregnancy complications.


Subject(s)
Cytomegalovirus Infections/genetics , Herpes Simplex/genetics , Pregnancy Complications, Infectious/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics , Adolescent , Adult , Alleles , Brazil , Case-Control Studies , Cytomegalovirus/immunology , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Epistasis, Genetic/immunology , Female , Genetic Predisposition to Disease , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/isolation & purification , Herpesvirus 2, Human/immunology , Herpesvirus 2, Human/isolation & purification , Humans , Placenta/immunology , Placenta/virology , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Retrospective Studies , Young Adult
20.
Viruses ; 13(2)2021 02 20.
Article in English | MEDLINE | ID: mdl-33672623

ABSTRACT

Zika virus (ZIKV) causes Congenital Zika Syndrome (CZS) in individuals exposed prenatally. Here, we investigated polymorphisms in VEGFA, PTGS2, NOS3, TNF, and NOS2 genes as risk factors to CZS. Forty children with CZS and forty-eight children who were in utero exposed to ZIKV infection, but born without congenital anomalies, were evaluated. Children with CZS were predominantly infected by ZIKV in the first trimester (p < 0.001) and had mothers with lower educational level (p < 0.001) and family income (p < 0.001). We found higher risk of CZS due the allele rs2297518[A] of NOS2 (OR = 2.28, CI 95% 1.17-4.50, p = 0.015). T allele and TT/CT genotypes of the TNF rs1799724 and haplotypes associated with higher expression of TNF were more prevalent in children with CZS and severe microcephaly (p = 0.029, p = 0.041 and p = 0.030, respectively). Our findings showed higher risk of CZS due ZIKV infection in the first trimester and suggested that polymorphisms in NOS2 and TNF genes affect the risk of CZS and severe microcephaly.


Subject(s)
Microcephaly/genetics , Nitric Oxide Synthase Type II/genetics , Tumor Necrosis Factor-alpha/genetics , Zika Virus Infection/genetics , Zika Virus/physiology , Adult , Alleles , Brazil , Case-Control Studies , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Infant , Male , Microcephaly/metabolism , Microcephaly/virology , Nitric Oxide Synthase Type II/metabolism , Polymorphism, Genetic , Pregnancy , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Pregnancy Trimester, First , Tumor Necrosis Factor-alpha/metabolism , Young Adult , Zika Virus/genetics , Zika Virus Infection/congenital , Zika Virus Infection/metabolism , Zika Virus Infection/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...