Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.199
Filter
1.
Front Immunol ; 15: 1423435, 2024.
Article in English | MEDLINE | ID: mdl-38994354

ABSTRACT

Introduction: Latent tuberculosis infection (LTBI) is a common coinfection in people living with HIV (PWH). How LTBI and HIV exposure in utero influence the development of infant humoral immunity is not well characterized. To address this question, we assessed the relationship between maternal humoral responses in pregnant women with HIV or with HIV/LTBI on humoral responses in infants to BCG vaccination and TB acquisition. Methods: Plasma samples were obtained from mother infant pairs during pregnancy (14-34 wks gestation) and in infants at 12 and 44 wks of age from the IMPAACT P1078 clinical trial. LTBI was established by Interferon gamma release assay (IGRA). Progression to active TB (ATB) disease was observed in 5 women at various times after giving birth. All infants were BCG vaccinated at birth and tested for IGRA at 44 weeks. Mtb (PPD, ESAT6/CFP10, Ag85A, LAM), HIV (GP120), and Influenza (HA) specific IgG, IgM, and IgA were measured in plasma samples using a bead based Luminex assay with Flexmap 3D. Results: In maternal plasma there were no differences in Mtb-specific antibodies or viral antibodies in relation to maternal IGRA status. ATB progressors showed increases in Mtb-specific antibodies at diagnosis compared to study entry. However, when compared to the non-progressors at entry, progressors had higher levels of Ag85A IgG and reduced ESAT6/CFP10 IgG and LAM IgG, IgM, and IgA1. All infants showed a decrease in IgG to viral antigens (HIV GP120 and HA) from 12 to 44 weeks attributed to waning of maternally transferred antibody titers. However, Mtb-specific (PPD, ESAT6/CFP10, Ag85A, and LAM) IgG and IgM increased from 12 to 44 weeks. HIV and HA IgG levels in maternal and 12-week infant plasma were highly correlated, and ESAT6/CFP10 IgG and LAM IgG showed a relationship between maternal and infant Abs. Finally, in the subset of infants that tested IGRA positive at 44 weeks, we observed a trend for lower LAM IgM compared to IGRA- infants at 44 weeks. Discussion: The results from our study raise the possibility that antibodies to LAM are associated with protection from progression to ATB and support further research into the development of humoral immunity against TB through infection or vaccination.


Subject(s)
Antibodies, Bacterial , HIV Infections , Immunity, Humoral , Latent Tuberculosis , Humans , Female , Latent Tuberculosis/immunology , HIV Infections/immunology , Pregnancy , Infant , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Adult , Mycobacterium tuberculosis/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/blood , BCG Vaccine/immunology , Infant, Newborn , Coinfection/immunology , Male , Prenatal Exposure Delayed Effects/immunology
2.
Vet Microbiol ; 295: 110154, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959808

ABSTRACT

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


Subject(s)
Disease Resistance , Fetus , Porcine Reproductive and Respiratory Syndrome , Transcriptome , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Disease Resistance/genetics , Disease Resistance/immunology , Pregnancy , Animals , Swine , Female , Fetus/immunology , Fetus/virology , Gene Expression Regulation/immunology , Myocardium/immunology , Liver/immunology , Disease Susceptibility/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/veterinary , Kidney/immunology
3.
J Med Virol ; 96(7): e29819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39030992

ABSTRACT

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.


Subject(s)
COVID-19 , Macrophages , Placenta , Pregnancy Complications, Infectious , SARS-CoV-2 , Humans , Female , Pregnancy , COVID-19/immunology , COVID-19/virology , Placenta/immunology , Placenta/virology , Macrophages/immunology , Macrophages/virology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , SARS-CoV-2/immunology , Adult , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Virus Internalization
4.
Front Immunol ; 15: 1418678, 2024.
Article in English | MEDLINE | ID: mdl-39021574

ABSTRACT

Background: Knowledge about SARS-CoV-2 antibody dynamics in neonates and direct comparisons with maternal antibody responses are not well established. This study aimed to characterize and directly compare the maternal and infant antibody response in a national birth cohort from the Faroe Islands. Methods: The levels of immunoglobulins (Ig) targeting the receptor binding domain (RBD) of the spike protein and the nucleocapsid protein (N protein) of SARS-CoV-2 were investigated in maternal blood and umbilical cord blood from neonates. The study included 537 neonates and 565 mothers from the Faroe Islands, and follow-up samples were collected 12 months after birth. Multiple linear regression models were used to assess associations of maternal parameters with maternal and neonatal Ig levels and pregnancy outcomes. Results: The finding showed that neonates acquired varying levels of SARS-CoV-2 antibodies through transplacental transfer, and the levels were significantly influenced by the mother's vaccination and infection status. The study also found that maternal vaccination and the presence of SARS-CoV-2 antibodies targeting spike RBD were associated with gestational age and APGAR scores. Furthermore, the anti-RBD and -N protein-specific antibody response dynamics during 12 months after birth exhibited differences between mothers and children. RBD and N protein responses were maintained at follow-up in the mother's cohort, while only the N protein response was maintained at follow-up in the children's cohort. Conclusion: In conclusion, SARS-CoV-2-specific immune responses in newborns rely on maternal immunity, while the persistence of SARS-CoV-2-specific Igs appears to be differently regulated between mothers and children. The study provides new insights into the dynamics of SARS-CoV-2-specific immune responses in newborns and underscores the nuanced relationship between maternal factors and neonatal humoral responses.


Subject(s)
Antibodies, Viral , COVID-19 , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Female , SARS-CoV-2/immunology , COVID-19/immunology , Pregnancy , Antibodies, Viral/blood , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Infant, Newborn , Coronavirus Nucleocapsid Proteins/immunology , Adult , Immunity, Maternally-Acquired , Infant , Male , Cohort Studies , Phosphoproteins/immunology , Pregnancy Complications, Infectious/immunology , Fetal Blood/immunology
5.
Front Immunol ; 15: 1430901, 2024.
Article in English | MEDLINE | ID: mdl-38947337

ABSTRACT

A maternal vaccine to protect newborns against invasive Streptococcus agalactiae infection is a developing medical need. The vaccine should be offered during the third trimester of pregnancy and induce strong immune responses and placental transfer of protective antibodies. Polysaccharide vaccines against S. agalactiae conjugated to protein carriers are in advanced stages of development. Additionally, protein-based vaccines are also in development, showing great promise as they can provide protection regardless of serotype. Furthermore, safety concerns regarding a new vaccine are the main barriers identified. Here, we present vaccines in development and identified safety, cost, and efficacy concerns, especially in high-need, low-income countries.


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Streptococcus agalactiae/immunology , Humans , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Streptococcal Infections/microbiology , Streptococcal Vaccines/immunology , Pregnancy , Female , Animals , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/prevention & control , Pregnancy Complications, Infectious/microbiology , Vaccine Development , Infant, Newborn , Antibodies, Bacterial/immunology
6.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892429

ABSTRACT

South Africa is the epicentre of the global HIV pandemic, with 13.9% of its population infected. Preeclampsia (PE), a hypertensive disorder of pregnancy, is often comorbid with HIV infection, leading to multi-organ dysfunction and convulsions. The exact pathophysiology of preeclampsia is triggered by an altered maternal immune response or defective development of maternal tolerance to the semi-allogenic foetus via the complement system. The complement system plays a vital role in the innate immune system, generating inflammation, mediating the clearance of microbes and injured tissue materials, and a mediator of adaptive immunity. Moreover, the complement system has a dual effect, of protecting the host against HIV infection and enhancing HIV infectivity. An upregulation of regulatory proteins has been implicated as an adaptive phenomenon in response to elevated complement-mediated cell lysis in HIV infection, further aggravated by preeclamptic complement activation. In light of the high prevalence of HIV infection and preeclampsia in South Africa, this review discusses the association of complement proteins and their role in the synergy of HIV infection and preeclampsia in South Africa. It aims to identify women at elevated risk, leading to early diagnosis and better management with targeted drug therapy, thereby improving the understanding of immunological dysregulation.


Subject(s)
Complement System Proteins , HIV Infections , Pre-Eclampsia , Humans , Pre-Eclampsia/immunology , Pre-Eclampsia/epidemiology , Pregnancy , HIV Infections/complications , HIV Infections/immunology , Female , Complement System Proteins/metabolism , Complement System Proteins/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/epidemiology , South Africa/epidemiology , Comorbidity , Complement Activation
7.
Sci Rep ; 14(1): 10813, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734805

ABSTRACT

To evaluate the development of neutralizing Anti-Spike Protein IgG (Anti-S-IgG) during twin pregnancies before conception vs. during pregnancy. In this prospective study, three blood samples were collected from pregnant women and subjected to anti-S-IgG immunodiagnostics. The patient's medical records, including vaccination and PCR test results, were collected from the hospital's electronic database. Age-matched non-pregnant women were used as a control group. We enrolled 83 women with twin pregnancies. 49 women were vaccinated before conception, 21 women were vaccinated during pregnancy, and 13 were not vaccinated. Of the 13 women who weren't vaccinated, three became positive during pregnancy, and all three were severely ill. By contrast, in women who were vaccinated during or before pregnancy, COVID-19 infection during pregnancy caused only mild symptoms. A ten-fold lower level of neutralizing Anti-S-IgG in the 3rd trimester was observed in healthy women who were vaccinated before conception and remained healthy until discharge from the hospital after delivery 1605 (IQR: 763-2410) compared to the healthy women who were vaccinated during pregnancy 152 AU/mL (IQR: 54-360). This difference was higher among women who were infected by COVID-19 (as verified by a positive PCR test). The third-trimester level of neutralizing Ant-S-IgG in the infected group was 4770 AU/mL (4760-6100) in infected women vaccinated before conception compared to those vaccinated during pregnancy who had 70 AU/mL (IQR: 20-170) (p < 0.001). In women vaccinated at 13-16 weeks gestation, neutralizing Anti-S-IgG at 20-22 weeks went up to 372 AU/mL (IQR: 120-1598) but rapidly dropped to 112 AU/mL (IQR: 54-357) at 28-30 weeks, (p < 0.001), a faster decline than in women vaccinated at a median 22 weeks before conception. Being infected by COVID-19 before conception was linked to having low Anti-S-IgG levels during pregnancy, whereas being infected by COVID-19 during pregnancy led to a very high response in the 3rd trimester. In twin pregnancies, significantly lower neutralizing Anti-S-IgG levels were observed in women vaccinated during pregnancy compared to those vaccinated before conception, whether infected or not infected by COVID-19. A full course of vaccination before conception is recommended.Trial registration. ClinicalTrials.gov Protocol Registration and Results System (PRS) Receipt Release Date: October 4, 2021. https://clinicaltrials.gov/ ID: NCT04595214.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunoglobulin G , Pregnancy, Twin , SARS-CoV-2 , Vaccination , Humans , Female , Pregnancy , Pregnancy, Twin/immunology , Adult , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Prospective Studies , SARS-CoV-2/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Pregnancy Complications, Infectious/prevention & control , Pregnancy Complications, Infectious/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology
8.
J Med Virol ; 96(5): e29639, 2024 May.
Article in English | MEDLINE | ID: mdl-38708824

ABSTRACT

Hepatitis E virus (HEV) infection in pregnant women is associated with a wide spectrum of adverse consequences for both mother and fetus. The high mortality in this population appears to be associated with hormonal changes and consequent immunological changes. This study conducted an analysis of immune responses in pregnant women infected with HEV manifesting varying severity. Data mining analysis of the GSE79197 was utilized to examine differentially biological functions in pregnant women with HEV infection (P-HEV) versus without HEV infection (P-nHEV), P-HEV progressing to ALF (P-ALF) versus P-HEV, and P-HEV versus non-pregnant women with HEV infection (nP-HEV). We found cellular response to interleukin and immune response-regulating signalings were activated in P-HEV compared with P-nHEV. However, there was a significant decrease of immune responses, such as T cell activation, leukocyte cell-cell adhesion, regulation of lymphocyte activation, and immune response-regulating signaling pathway in P-ALF patient than P-HEV patient. Compared with nP-HEV, MHC protein complex binding function was inhibited in P-HEV. Further microRNA enrichment analysis showed that MAPK and T cell receptor signaling pathways were inhibited in P-HEV compared with nP-HEV. In summary, immune responses were activated during HEV infection while being suppressed when developing ALF during pregnancy, heightening the importance of immune mediation in the pathogenesis of severe outcome in HEV infected pregnant women.


Subject(s)
Hepatitis E virus , Hepatitis E , Pregnancy Complications, Infectious , Humans , Female , Pregnancy , Hepatitis E/immunology , Hepatitis E/virology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , Hepatitis E virus/immunology , Signal Transduction , Liver Failure, Acute/immunology , Liver Failure, Acute/virology , MicroRNAs/genetics , Adult
9.
BMC Infect Dis ; 24(1): 509, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773493

ABSTRACT

PURPOSE: Pregnant women are at risk of severe SARS-CoV-2 infection, potentially leading to obstetric and neonatal complications. Placental transfer of antibodies directed to SARS-CoV-2 may be protective against neonatal COVID-19, but this remains to be studied. We aimed to determine the seroprevalence of SARS-CoV-2 antibodies in a population of unvaccinated pregnant women and to determine the placental transfer of these antibodies. METHODOLOGY: A total of 1197 unvaccinated women with mostly unknown pre-study SARS-CoV-2 infection status, were tested at delivery for SARS-CoV-2 spike protein IgG antibodies during the first year of the pandemic. Umbilical cord samples were collected and assessed for seropositivity if the mother was seropositive. Maternal characteristics, pregnancy and neonatal outcomes and data on SARS-CoV-2 infection were extracted from medical records. RESULTS: Specific IgG were detected in 258 women (21.6%). A significant placental transfer to the newborn was observed in 81.3% of cases. The earlier in the 2nd and 3rd trimesters that the mother had contracted the disease and the more symptomatic she was, the greater the likelihood of transplacental transfer of IgG to her newborn. CONCLUSION: Approximately one in five women had detectable anti-SARS-CoV-2 spike protein IgG antibodies at delivery during the first year of the pandemic, and these antibodies were significantly transferred to their fetuses. This research provides further evidence to better understand the dynamics of the placental transfer of SARS-CoV-2 IgG antibodies from mothers to their newborns, which is necessary to improve vaccination strategies.


Subject(s)
Antibodies, Viral , COVID-19 , Immunoglobulin G , Pregnancy Complications, Infectious , SARS-CoV-2 , Humans , Female , Pregnancy , COVID-19/immunology , COVID-19/epidemiology , Seroepidemiologic Studies , SARS-CoV-2/immunology , Adult , Antibodies, Viral/blood , Immunoglobulin G/blood , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/immunology , Infant, Newborn , Spike Glycoprotein, Coronavirus/immunology , Placenta/immunology , Young Adult , Infectious Disease Transmission, Vertical , Maternal-Fetal Exchange/immunology
10.
Viruses ; 16(5)2024 05 14.
Article in English | MEDLINE | ID: mdl-38793661

ABSTRACT

Human cytomegalovirus (CMV) is a common herpesvirus causing lifelong latent infection in most people and is a primary cause of congenital infection worldwide. Given the role of NK cells in the materno-fetal barrier, we investigated peripheral blood NK cell behavior in the context of CMV infection acquired during pregnancy. We analyzed the NK phenotype and CD107a surface mobilization on PBMCs from CMV-transmitting and non-transmitting mothers and newborns with or without congenital infection. NK cells from non-transmitting mothers showed the typical phenotype of CMV-adaptive NK cells, characterized by higher levels of NKG2C, CD57, and KIRs, with reduced NKG2A, compared to transmitting ones. A significantly higher percentage of DNAM-1+, PD-1+, and KIR+NKG2A-CD57+PD-1+ CD56dim cells was found in the non-transmitting group. Accordingly, NK cells from congenital-CMV (cCMV)-infected newborns expressed higher levels of NKG2C and CD57, with reduced NKG2A, compared to non-congenital ones. Furthermore, they showed a significant expansion of CD56dim cells co-expressing NKG2C and CD57 or with a memory-like (KIR+NKG2A-CD57+NKG2C+) phenotype, as well as a significant reduction of the CD57-NKG2C- population. Degranulation assays showed a slightly higher CD107a geomean ratio in NK cells of mothers who were non-transmitting compared to those transmitting the virus. Our findings demonstrate that both CMV-transmitting mothers and cCMV newborns show a specific NK profile. These data can guide studies on predicting virus transmission from mothers and congenital infection in infants.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Infectious Disease Transmission, Vertical , Killer Cells, Natural , Pregnancy Complications, Infectious , Humans , Killer Cells, Natural/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/transmission , Female , Pregnancy , Infant, Newborn , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , Cytomegalovirus/immunology , Adult , Cohort Studies , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Young Adult
11.
Sci Rep ; 14(1): 10980, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744864

ABSTRACT

During pregnancy, multiple immune regulatory mechanisms establish an immune-tolerant environment for the allogeneic fetus, including cellular signals called cytokines that modify immune responses. However, the impact of maternal HIV infection on these responses is incompletely characterized. We analyzed paired maternal and umbilical cord plasma collected during labor from 147 people with HIV taking antiretroviral therapy and 142 HIV-uninfected comparators. Though cytokine concentrations were overall similar between groups, using Partial Least Squares Discriminant Analysis we identified distinct cytokine profiles in each group, driven by higher IL-5 and lower IL-8 and MIP-1α levels in pregnant people with HIV and higher RANTES and E-selectin in HIV-unexposed umbilical cord plasma (P-value < 0.01). Furthermore, maternal RANTES, SDF-α, gro α -KC, IL-6, and IP-10 levels differed significantly by HIV serostatus (P < 0.01). Although global maternal and umbilical cord cytokine profiles differed significantly (P < 0.01), umbilical cord plasma profiles were similar by maternal HIV serostatus. We demonstrate that HIV infection is associated with a distinct maternal plasma cytokine profile which is not transferred across the placenta, indicating a placental role in coordinating local inflammatory response. Furthermore, maternal cytokine profiles in people with HIV suggest an incomplete shift from Th2 to Th1 immune phenotype at the end of pregnancy.


Subject(s)
Cytokines , HIV Infections , Pregnancy Complications, Infectious , Humans , Pregnancy , Female , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , Cytokines/blood , Adult , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Uganda , Fetal Blood/metabolism , Young Adult
12.
Hum Antibodies ; 32(2): 61-65, 2024.
Article in English | MEDLINE | ID: mdl-38669524

ABSTRACT

BACKGROUND: The antibody that crosses transplacentally from mother to fetus is very important origin of protective passive immunity against infection neonatal with enterovirus. Important varieties of coxsackievirus B3 (CVB3) are responsible for infections in newborns. The purpose from this study is to investigate in the prevalence of Coxsackie B virus in a sample of Iraqi women with miscarriage and potential role of miscarriage risk. METHODS: Between November 2022 and June 2023, we included 91 parturient women (gestational age: 4-20 weeks) who were between the ages of 15 and 40. Every participant completed a questionnaire, and blood was drawn to assess maternal antibodies against CVB3. RESULTS: The blood seropositive rates were 46 out 91(50.54%), 2 out 46 were IgM positive (4.34%), (8-12 weeks) 23 from 46 (50%) (p-value 0.0294) gestational age more frequent among aborted women that positive for anti-coxsackie B antibody, The 25-35 age group was significantly overrepresented (51/91, 56%) compared to other age groups. CONCLUSION: This investigation posits Coxsackie B virus (CBV) as a possible etiology for miscarriage in the Iraqi female population. Further studies employing larger cohorts and robust methodologies, beyond the current detection technique, are warranted to corroborate these observations and elucidate the potential mechanisms by which CBV might induce miscarriage.


Subject(s)
Abortion, Spontaneous , Antibodies, Viral , Coxsackievirus Infections , Enterovirus B, Human , Humans , Female , Iraq/epidemiology , Adult , Pregnancy , Enterovirus B, Human/immunology , Abortion, Spontaneous/virology , Abortion, Spontaneous/immunology , Abortion, Spontaneous/epidemiology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/epidemiology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Young Adult , Adolescent , Immunoglobulin M/blood , Immunoglobulin M/immunology , Seroepidemiologic Studies , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/epidemiology , Prevalence
13.
Biomolecules ; 14(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38672452

ABSTRACT

Vaccination against SARS-CoV-2 has been demonstrated to be safe during gestation. Nevertheless, there are no robust data investigating the entity of maternal antibodies' transmission through the placenta to the newborn and the persistence of the antibodies in babies' serum. The objective of this study is to assess the maternal antibody transmission and kinetics among newborns in the first months of life. Women having received one or two doses of anti-SARS-CoV-2 mRNA-vaccines during pregnancy at any gestational age, and their newborns, were recruited and followed-up over 9 months. Ninety-eight women and 103 babies were included. At birth, we observed a significant positive correlation between maternal and neonatal serum anti-SARS-CoV-2 antibody levels and a significant negative correlation between the time since last dose and antibody levels in mothers with two doses. Over the follow-up, the birth antibody level significantly decreased in time according to the received doses number at 3, 6, and 9 months. During the follow-up, we registered 34 dyad SARS-CoV-2 infection cases. The decreasing trend was slower in the SARS-CoV-2 infection group and among breastfed non-infected babies. Antibodies from maternal anti-SARS-CoV-2 vaccination are efficiently transferred via the placenta and potentially even through breast milk. Among newborns, antibodies show relevant durability in the first months of life.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Humans , Female , Pregnancy , COVID-19/immunology , COVID-19/prevention & control , Infant, Newborn , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Adult , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccination , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Infant
14.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673883

ABSTRACT

Respiratory infections are common in pregnancy with conflicting evidence supporting their association with neonatal congenital anomalies, especially during the first trimester. We profiled cytokine and chemokine systemic responses in 242 pregnant women and their newborns after SARS-CoV-2 infection, acquired in different trimesters. Also, we tested transplacental IgG passage and maternal vaginal-rectal microbiomes. IgG transplacental passage was evident, especially with infection acquired in the first trimester. G-CSF concentration-involved in immune cell recruitment-decreased in infected women compared to uninfected ones: a beneficial event for the reduction of inflammation but detrimental to ability to fight infections at birth. The later the infection was acquired, the higher the systemic concentration of IL-8, IP-10, and MCP-1, associated with COVID-19 disease severity. All infected women showed dysbiosis of vaginal and rectal microbiomes, compared to uninfected ones. Two newborns tested positive for SARS-CoV-2 within the first 48 h of life. Notably, their mothers had acute infection at delivery. Although respiratory infections in pregnancy are reported to affect babies' health, with SARS-CoV-2 acquired early during gestation this risk seems low because of the maternal immune response. The observed vaginal and rectal dysbiosis could be relevant for neonatal microbiome establishment, although in our series immediate neonatal outcomes were reassuring.


Subject(s)
COVID-19 , Dysbiosis , Pregnancy Complications, Infectious , SARS-CoV-2 , Vagina , Humans , Female , Pregnancy , COVID-19/immunology , Dysbiosis/immunology , Dysbiosis/microbiology , Adult , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/virology , Vagina/microbiology , Vagina/immunology , Vagina/virology , Infant, Newborn , Cytokines/metabolism , Pregnancy Trimesters/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Microbiota/immunology
15.
Immun Inflamm Dis ; 12(4): e1257, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661110

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is the world's leading cause of viral acute lower respiratory infections (ALRI) in infants. WHO has identified maternal RSV vaccination a priority and candidate vaccines are in development; however, vaccine hesitancy remains an impediment to successful implementation of maternal immunization. This study, the largest antenatal survey conducted to-date, aimed to examine maternal RSV awareness, likely acceptance of RSV vaccination in pregnancy, and attitudes to maternal vaccination. METHODS: Pregnant women of all gestations attending antenatal clinic of a university maternity hospital in Ireland were invited to participate. An information leaflet provided, consent obtained, and survey administered examining RSV awareness, willingness to avail of antenatal RSV vaccination, factors influencing acceptability and preferred sources of assistance. Research Ethics Committee (REC) approval obtained, and general data protection regulation (GDPR) guidelines followed. RESULTS: 528 women completed the survey. A large proportion (75.6%) had never heard of RSV, yet 48.5% would still avail of a vaccine, 45.8% were undecided and only 5.3% would not. The main factor making vaccination acceptable to women (76.4%) was that it protects their infant from illness (p < .001, CV 0.336 for association with acceptance) and general practitioner (GP) was the preferred guidance source in decision-making (57.7%). CONCLUSIONS: Despite low levels of maternal awareness of RSV, pregnant women in Ireland are open to availing of antenatal vaccination. Maternal immunization strategies need to focus on infant's protection from RSV-associated ALRI along with vaccine safety, and build on an interdisciplinary collaboration of maternal, neonatal, primary care and public health services.


Subject(s)
Health Knowledge, Attitudes, Practice , Patient Acceptance of Health Care , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Vaccination , Humans , Female , Ireland/epidemiology , Pregnancy , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/psychology , Respiratory Syncytial Virus Infections/immunology , Adult , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Vaccination/psychology , Patient Acceptance of Health Care/psychology , Patient Acceptance of Health Care/statistics & numerical data , Pregnancy Complications, Infectious/prevention & control , Pregnancy Complications, Infectious/psychology , Pregnancy Complications, Infectious/immunology , Surveys and Questionnaires , Young Adult , Vaccination Hesitancy/psychology , Vaccination Hesitancy/statistics & numerical data , Pregnant Women/psychology , Respiratory Syncytial Virus, Human/immunology , Adolescent
16.
J Reprod Immunol ; 163: 104220, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38447289

ABSTRACT

Tumor necrosis factor alpha (TNFα) is involved in the occurrence of negative pregnancy outcomes. The study aimed to evaluate the safety and efficacy of the immunosuppressive TNFα inhibitors (TNFαi) in the treatment of patients with a history of recurrent reproductive failure in the context of COVID-19 pandemics. We reviewed 85 patients who received TNFαi (certolizumab pegol) during Mainland China's first wave of COVID-19 pandemic, from 21st Nov 2022-11 th Jan 2023. We also collected corresponding data from 130 pregnant patients who never used TNFαi for comparison. There were no significant differences in the history of previous pregnancy loss, miscarriage, embryo implantation failure, comorbidities and doses of COVID-19 vaccination. 82.2% and 87.7% pregnant patients contracted primary COVID-19 with symptoms in TNFαi group and no-TNFαi group. Duration of symptoms was significantly longer in TNFαi group and the incidences of cough and lethargy was significantly higher in TNFαi group. Both groups reported similar severity to same-aged close contacts, similar rates of other symptoms and hospitalization. No deaths were reported. In the in vitro fertilization (IVF) subgroup, we achieved a biochemical pregnancy loss rate of 17.4%, miscarriage rate of 21.7%, ongoing pregnancy rate and live birth rate of 34.2%. COVID-19 did not influence the live birth rate. We concluded that TNFαi administration in pregnancy was not associated with increased susceptivity to and severity of COVID-19. However, TNFαi users showed more prominent symptoms and longer recovery time. The pregnancy outcomes with TNFαi in such high-risk group for pregnancy loss was satisfactory.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , SARS-CoV-2 , Tumor Necrosis Factor-alpha , Humans , Pregnancy , Female , COVID-19/epidemiology , COVID-19/immunology , SARS-CoV-2/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/virology , Adult , Pregnancy Outcome , China/epidemiology , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor Inhibitors/adverse effects , Retrospective Studies
17.
Protein Cell ; 15(6): 460-473, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38441496

ABSTRACT

The current coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2) remains a threat to pregnant women. However, the impact of early pregnancy SARS-CoV-2 infection on the maternal-fetal interface remains poorly understood. Here, we present a comprehensive analysis of single-cell transcriptomics and metabolomics in placental samples infected with SARS-CoV-2 during early pregnancy. Compared to control placentas, SARS-CoV-2 infection elicited immune responses at the maternal-fetal interface and induced metabolic alterations in amino acid and phospholipid profiles during the initial weeks post-infection. However, subsequent immune cell activation and heightened immune tolerance in trophoblast cells established a novel dynamic equilibrium that mitigated the impact on the maternal-fetal interface. Notably, the immune response and metabolic alterations at the maternal-fetal interface exhibited a gradual decline during the second trimester. Our study underscores the adaptive immune tolerance mechanisms and establishment of immunological balance during the first two trimesters following maternal SARS-CoV-2 infection.


Subject(s)
COVID-19 , Placenta , Pregnancy Complications, Infectious , SARS-CoV-2 , Female , Pregnancy , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Placenta/immunology , Placenta/virology , Placenta/metabolism , Immune Tolerance , Trophoblasts/immunology , Trophoblasts/metabolism , Trophoblasts/virology , Adult , Pregnancy Trimester, First/immunology , Transcriptome
18.
J Reprod Immunol ; 163: 104243, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522364

ABSTRACT

Associations between antenatal SARS-CoV-2 infection and pregnancy outcomes have been conflicting and the role of the immune system is currently unclear. This prospective cohort study investigated the interaction of antenatal SARS-CoV-2 infection, changes in cytokine and HS-CRP levels, birthweight and gestational age at birth. 2352 pregnant participants from New York City (2020-2022) were included. Plasma levels of interleukin (IL)-1ß, IL-6, IL-17A and high-sensitivity C-reactive protein (HS-CRP) were quantified in blood specimens obtained across pregnancy. Quantile and linear regression models were conducted to 1) assess the impact of antenatal SARS-CoV-2 infection, overall and by timing of detection of SARS-CoV-2 positivity (< 20 weeks versus ≥ 20 weeks), on birthweight and gestational age at delivery; 2) examine the relationship between SARS-CoV-2 infection and maternal immune changes during pregnancy. All models were adjusted for maternal demographic and obstetric factors and pandemic timing. Birthweight models were additionally adjusted for gestational age at delivery and fetal sex. Immune marker models were also adjusted for gestational age at specimen collection and multiplex assay batch. 371 (15.8%) participants were infected with SARS-CoV-2 during pregnancy, of which 98 (26.4%) were infected at < 20 weeks gestation. Neither SARS-CoV-2 infection in general nor in early or late pregnancy was associated with lower birthweight nor earlier gestational age at delivery. Further, we did not observe cytokine or HS-CRP changes in response to SARS-CoV-2 infection and thus found no evidence to support a potential association between immune dysregulation and the diversity in pregnancy outcomes following infection.


Subject(s)
Birth Weight , COVID-19 , Inflammation , Pregnancy Complications, Infectious , Pregnancy Outcome , SARS-CoV-2 , Humans , Pregnancy , Female , COVID-19/immunology , COVID-19/blood , Adult , Prospective Studies , New York City/epidemiology , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/virology , Inflammation/immunology , Inflammation/blood , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Gestational Age , Infant, Newborn , Cytokines/blood
19.
AIDS ; 38(7): 945-954, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38329228

ABSTRACT

OBJECTIVE: CCR5-tropic viruses are preferentially transmitted during perinatal HIV-1 infection. CCR5 density on CD4 + T-cells likely impacts susceptibility to HIV-1 infection. DESIGN: Fifty-two mother-infant dyads were enrolled. All mothers were living with HIV-1, 27 of the infants acquired HIV-1 in utero and 25 infants remained uninfected. METHODS: CCR5 density, together with frequencies of CD4 + and CD8 + T-cells expressing immune activation (CCR5, ICOS and HLA-DR) and immune checkpoint (TIGIT and PD-1) markers, were measured in whole blood from the dyads close to delivery. RESULTS: Compared with mothers who did not transmit, mothers who transmitted HIV-1 had less exposure to ART during pregnancy ( P = 0.015) and higher plasma viral load close to delivery ( P = 0.0005). These mothers, additionally, had higher CCR5 density on CD4 + and CD8 + T-cells and higher frequencies of CCR5, ICOS and TIGIT-expressing CD8 + T-cells. Similarly, compared with infants without HIV-1, infants with HIV-1 had higher CCR5 density on CD4 + and CD8 + T-cells and higher frequencies of CCR5, TIGIT, and PD-1-expressing CD4 + and CD8 + T-cells as well as higher frequencies of HLA-DR-expressing CD8 + T-cells. CCR5 density on maternal CD4 + T-cells remained significantly associated with transmission after adjusting for maternal viral load and CD4 + T cell counts. Mother-infant dyads with shared high CCR5 density phenotypes had the highest risk of transmission/acquisition of infection compared with dyads with shared low-CCR5 density phenotypes. CONCLUSION: This study provides strong evidence of a protective role for a combined mother-infant low CD4 + T-cell CCR5 density phenotype in in-utero transmission/acquisition of HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes , HIV Infections , HIV-1 , Infectious Disease Transmission, Vertical , Receptors, CCR5 , Humans , Female , HIV Infections/transmission , HIV Infections/immunology , Pregnancy , CD4-Positive T-Lymphocytes/immunology , Adult , HIV-1/immunology , Infant, Newborn , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , Infant , Male , Young Adult , Viral Load , Risk Assessment , CD8-Positive T-Lymphocytes/immunology
20.
Clin Microbiol Rev ; 37(2): e0007323, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38421182

ABSTRACT

SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring.


Subject(s)
Pregnancy Complications, Infectious , Virus Diseases , Pregnancy , Female , Humans , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Animals , Virus Diseases/immunology , Virus Diseases/transmission , Placenta/virology , Placenta/immunology , Infectious Disease Transmission, Vertical , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL