Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.239
Filtrer
1.
J Clin Invest ; 134(12)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38950310

RÉSUMÉ

In utero gene editing (IUGE) is a potential treatment for inherited diseases that cause pathology before or soon after birth. Preexisting immunity to adeno-associated virus (AAV) vectors and Cas9 endonuclease may limit postnatal gene editing. The tolerogenic fetal immune system minimizes a fetal immune barrier to IUGE. However, the ability of maternal immunity to limit fetal gene editing remains a question. We investigated whether preexisting maternal immunity to AAV or Cas9 impairs IUGE. Using a combination of fluorescent reporter mice and a murine model of a metabolic liver disease, we demonstrated that maternal anti-AAV IgG antibodies were efficiently transferred from dam to fetus and impaired IUGE in a maternal titer-dependent fashion. By contrast, maternal cellular immunity was inefficiently transferred to the fetus, and neither maternal cellular nor humoral immunity to Cas9 impaired IUGE. Using human umbilical cord and maternal blood samples collected from mid- to late-gestation pregnancies, we demonstrated that maternal-fetal transmission of anti-AAV IgG was inefficient in midgestation compared with term, suggesting that the maternal immune barrier to clinical IUGE would be less relevant at midgestation. These findings support immunologic advantages for IUGE and inform maternal preprocedural testing protocols and exclusion criteria for future clinical trials.


Sujet(s)
Dependovirus , Édition de gène , Animaux , Femelle , Dependovirus/génétique , Dependovirus/immunologie , Souris , Grossesse , Humains , Immunoglobuline G/immunologie , Immunoglobuline G/génétique , Immunoglobuline G/sang , Protéine-9 associée à CRISPR/génétique , Protéine-9 associée à CRISPR/immunologie , Vecteurs génétiques/immunologie , Échange foetomaternel/immunologie , Échange foetomaternel/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Systèmes CRISPR-Cas , Foetus/immunologie , Immunité acquise d'origine maternelle/immunologie
2.
Front Immunol ; 15: 1415794, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957469

RÉSUMÉ

Endocytosis represents a category of regulated active transport mechanisms. These encompass clathrin-dependent and -independent mechanisms, as well as fluid phase micropinocytosis and macropinocytosis, each demonstrating varying degrees of specificity and capacity. Collectively, these mechanisms facilitate the internalization of cargo into cellular vesicles. Pregnancy is one such physiological state during which endocytosis may play critical roles. A successful pregnancy necessitates ongoing communication between maternal and fetal cells at the maternal-fetal interface to ensure immunologic tolerance for the semi-allogenic fetus whilst providing adequate protection against infection from pathogens, such as viruses and bacteria. It also requires transport of nutrients across the maternal-fetal interface, but restriction of potentially harmful chemicals and drugs to allow fetal development. In this context, trogocytosis, a specific form of endocytosis, plays a crucial role in immunological tolerance and infection prevention. Endocytosis is also thought to play a significant role in nutrient and toxin handling at the maternal-fetal interface, though its mechanisms remain less understood. A comprehensive understanding of endocytosis and its mechanisms not only enhances our knowledge of maternal-fetal interactions but is also essential for identifying the pathogenesis of pregnancy pathologies and providing new avenues for therapeutic intervention.


Sujet(s)
Endocytose , Échange foetomaternel , Humains , Grossesse , Endocytose/immunologie , Femelle , Échange foetomaternel/immunologie , Animaux , Transport biologique , Nutriments/métabolisme , Tolérance immunitaire , Placenta/immunologie , Placenta/métabolisme
3.
Physiol Res ; 73(3): 315-332, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39027950

RÉSUMÉ

Maternal-fetal microchimerism is a fascinating phenomenon in which maternal cells migrate to the tissues of the offspring during both pregnancy and breastfeeding. These cells primarily consist of leukocytes and stem cells. Remarkably, these maternal cells possess functional potential in the offspring and play a significant role in shaping their immune system development. T lymphocytes, a cell population mainly found in various tissues of the offspring, have been identified as the major cell type derived from maternal microchimerism. These T lymphocytes not only exert effector functions but also influence the development of the offspring's T lymphocytes in the thymus and the maturation of B lymphocytes in the lymph nodes. Furthermore, the migration of maternal leukocytes also facilitates the transfer of immune memory across generations. Maternal microchimerism has also been observed to address immunodeficiencies in the offspring. This review article focuses on investigating the impact of maternal cells transported within maternal microchimerism on the immune system development of the offspring, as well as elucidating the effector functions of maternal cells that migrate through the placenta and breast milk to reach the offspring.


Sujet(s)
Chimérisme , Mémoire immunologique , Échange foetomaternel , Humains , Femelle , Grossesse , Échange foetomaternel/immunologie , Animaux
4.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830841

RÉSUMÉ

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Sujet(s)
Foetus , Lipopolysaccharides , Foie , Poumon , Placenta , Femelle , Grossesse , Placenta/métabolisme , Placenta/immunologie , Animaux , Foetus/immunologie , Foetus/métabolisme , Poumon/immunologie , Poumon/métabolisme , Foie/métabolisme , Foie/immunologie , Acide docosahexaénoïque/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Souris , Inflammation/immunologie , Inflammation/métabolisme , Souris de lignée C57BL , Adaptation physiologique/immunologie , Développement foetal/immunologie , Échange foetomaternel/immunologie , Interleukine-6/métabolisme , Interleukine-6/immunologie
5.
Chin Med J (Engl) ; 137(12): 1399-1406, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38724467

RÉSUMÉ

ABSTRACT: Normal pregnancy is a contradictory and complicated physiological process. Although the fetus carries the human leukocyte antigen (HLA) inherited from the paternal line, it does not cause maternal immune rejection. As the only exception to immunological principles, maternal-fetal immune tolerance has been a reproductive immunology focus. In early pregnancy, fetal extravillous trophoblast cells (EVTs) invade decidual tissues and come into direct contact with maternal decidual immune cells (DICs) and decidual stromal cells (DSCs) to establish a sophisticated maternal-fetal crosstalk. This study reviews previous research results and focuses on the establishment and maintenance mechanism of maternal-fetal tolerance based on maternal-fetal crosstalk. Insights into maternal-fetal tolerance will not only improve understanding of normal pregnancy but will also contribute to novel therapeutic strategies for recurrent spontaneous abortion, pre-eclampsia, and premature birth.


Sujet(s)
Tolérance immunitaire , Humains , Grossesse , Femelle , Tolérance immunitaire/immunologie , Échange foetomaternel/immunologie , Caduques/immunologie , Trophoblastes/immunologie , Foetus/immunologie
6.
Am J Reprod Immunol ; 91(5): e13859, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38722063

RÉSUMÉ

Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.


Sujet(s)
Avortements à répétition , Humains , Femelle , Grossesse , Avortements à répétition/immunologie , Tolérance immunitaire , Échange foetomaternel/immunologie , Microbiome gastro-intestinal/immunologie , Immunothérapie/méthodes
7.
Article de Anglais | MEDLINE | ID: mdl-38782369

RÉSUMÉ

Pregnancy is a remarkable event where the semi-allogeneic fetus develops in the mother's uterus, despite genetic and immunological differences. The antigen handling and processing at the maternal-fetal interface during pregnancy appear to be crucial for the adaptation of the maternal immune system and for tolerance to the developing fetus and placenta. Maternal antigen-presenting cells (APCs), such as macrophages (Mφs) and dendritic cells (DCs), are present at the maternal-fetal interface throughout pregnancy and are believed to play a crucial role in this process. Despite numerous studies focusing on the significance of Mφs, there is limited knowledge regarding the contribution of DCs in fetomaternal tolerance during pregnancy, making it a relatively new and growing field of research. This review focuses on how the behavior of DCs at the maternal-fetal interface adapts to pregnancy's unique demands. Moreover, it discusses how DCs interact with other cells in the decidual leukocyte network to regulate uterine and placental homeostasis and the local maternal immune responses to the fetus. The review particularly examines the different cell lineages of DCs with specific surface markers, which have not been critically reviewed in previous publications. Additionally, it emphasizes the impact that even minor disruptions in DC functions can have on pregnancy-related complications and proposes further research into the potential therapeutic benefits of targeting DCs to manage these complications.


Sujet(s)
Cellules dendritiques , Tolérance immunitaire , Échange foetomaternel , Placenta , Humains , Grossesse , Cellules dendritiques/immunologie , Femelle , Échange foetomaternel/immunologie , Placenta/immunologie , Foetus/immunologie , Animaux , Macrophages/immunologie , Complications de la grossesse/immunologie
8.
BMC Infect Dis ; 24(1): 509, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773493

RÉSUMÉ

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.


Sujet(s)
Anticorps antiviraux , COVID-19 , Immunoglobuline G , Complications infectieuses de la grossesse , SARS-CoV-2 , Humains , Femelle , Grossesse , COVID-19/immunologie , COVID-19/épidémiologie , Études séroépidémiologiques , SARS-CoV-2/immunologie , Adulte , Anticorps antiviraux/sang , Immunoglobuline G/sang , Complications infectieuses de la grossesse/épidémiologie , Complications infectieuses de la grossesse/immunologie , Nouveau-né , Glycoprotéine de spicule des coronavirus/immunologie , Placenta/immunologie , Jeune adulte , Transmission verticale de maladie infectieuse , Échange foetomaternel/immunologie
9.
Sci Rep ; 14(1): 10772, 2024 05 10.
Article de Anglais | MEDLINE | ID: mdl-38730052

RÉSUMÉ

We aimed to determine SARS-CoV-2 antibody seropositivity among pregnant women and the transplacental transfer efficiency of SARS-CoV-2-specific antibodies relative to malaria antibodies among SARS-CoV-2 seropositive mother-cord pairs. This cross-sectional study was conducted in Accra, Ghana, from March to May 2022. Antigen- specific IgG antibodies against SARS-CoV-2 (nucleoprotein and spike-receptor binding domain) and malarial antigens (circumsporozoite protein and merozoite surface protein 3) in maternal and cord plasma were measured by ELISA. Plasma from both vaccinated and unvaccinated pregnant women were tested for neutralizing antibodies using commercial kit. Of the unvaccinated pregnant women tested, 58.12% at antenatal clinics and 55.56% at the delivery wards were seropositive for both SARS-CoV-2 nucleoprotein and RBD antibodies. Anti-SARS-CoV-2 antibodies in cord samples correlated with maternal antibody levels (N antigen rs = 0.7155, p < 0.001; RBD rs = 0.8693, p < 0.001). Transplacental transfer of SARS-CoV-2 nucleoprotein antibodies was comparable to circumsporozoite protein antibodies (p = 0.9999) but both were higher than transfer rates of merozoite surface protein 3 antibodies (p < 0.001). SARS-CoV-2 IgG seropositivity among pregnant women in Accra is high with a boost of SARS-CoV-2 RBD-specific IgG in vaccinated women. Transplacental transfer of anti-SARS-CoV-2 and malarial antibodies was efficient, supporting vaccination of mothers as a strategy to protect infants against SARS-CoV-2.


Sujet(s)
Anticorps antiviraux , COVID-19 , Immunoglobuline G , SARS-CoV-2 , Humains , Femelle , Grossesse , Ghana , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Adulte , Études transversales , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Échange foetomaternel/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Nourrisson , Nouveau-né , Glycoprotéine de spicule des coronavirus/immunologie , Immunité acquise d'origine maternelle , Jeune adulte , Sang foetal/immunologie , Anticorps antiprotozoaires/immunologie , Anticorps antiprotozoaires/sang
10.
Int J Infect Dis ; 144: 107047, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38609035

RÉSUMÉ

AIM/OBJECTIVE: This study investigates placental antibody transfer following recombinant pertussis vaccination in pregnancy in a real-world setting. METHODS: This postmarketing observational study recruited pregnant women vaccinated with monovalent recombinant acellular pertussis (aP) vaccine (aPgen; n = 199) or combined to tetanus-diphtheria (TdaPgen; n = 200), or Td-vaccine only (n = 54). Pregnancy, delivery, and neonatal outcomes were assessed. Cord blood was collected postdelivery and pertussis toxin (PT)-IgG, filamentous hemagglutinin (FHA)-IgG, and PT-neutralizing antibodies (PT-Nab) were assessed. RESULTS: No adverse pregnancy, delivery, or neonatal outcomes attributed to aPgen, TdaPgen, or Td vaccination were reported. High anti-PT antibody levels were detected in cord samples from women vaccinated with aPgen (geometric mean concentration [GMC] PT-IgG 206.1 IU/ml, 95% confidence intervals [CI]: 164.3-258.6; geometric mean titer [GMT] PT-Nab 105.3 IU/ml, 95% CI: 81.7-135.8) or TdaPgen (GMC PT-IgG 153.1 IU/ml, 95% CI: 129.1-181.5; GMT PT-Nab 81.5 IU/ml, 95% CI: 66.4-100.0). In the Td-only group, anti-PT antibodies were low (GMC PT-IgG 6.5 IU/ml, 95% CI: 4.9-8.8; GMT PT-Nab 3.8 IU/ml, 95% CI: 2.8-5.1). The same was found for FHA-IgG. Recombinant pertussis vaccination at <27 or 27-36 weeks gestation induced similar cord pertussis antibody levels. CONCLUSION: This first real-world study confirms that recombinant pertussis vaccination in the second or third trimester of pregnancy results in high levels of passive immunity in infants. Thai Clinical Trial Registry: TCTR20200528006.


Sujet(s)
Anticorps antibactériens , Immunité acquise d'origine maternelle , Coqueluche , Humains , Femelle , Grossesse , Adulte , Anticorps antibactériens/sang , Anticorps antibactériens/immunologie , Coqueluche/prévention et contrôle , Coqueluche/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Sang foetal/immunologie , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Vaccin anticoquelucheux/immunologie , Vaccin anticoquelucheux/administration et posologie , Jeune adulte , Échange foetomaternel/immunologie , Vaccins diphtérique tétanique coquelucheux acellulaires/immunologie , Vaccins diphtérique tétanique coquelucheux acellulaires/administration et posologie , Nouveau-né , Toxine pertussique/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Bordetella pertussis/immunologie , Vaccination
11.
J Reprod Immunol ; 163: 104244, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38555747

RÉSUMÉ

Implantation and maintenance of pregnancy involve intricate immunological processes that enable the developing fetus to coexist with the maternal immune system. Progesterone, a critical hormone during pregnancy, is known to promote immune tolerance and prevent preterm labor. However, the mechanism by which progesterone mediates these effects remains unclear. In this study, we investigated the role of the non-classical progesterone receptor membrane component 1 (PGRMC1) in progesterone signaling at the maternal-fetal interface. Using JEG3 cells, a trophoblast model cell line, we observed that progesterone stimulation increased the expression of human leukocyte antigen-C (HLA-C) and HLA-G, key molecules involved in immune tolerance. We also found that progesterone upregulated the expression of the transcription factor ELF3, which is known to regulate trophoblast-specific HLA-C expression. Interestingly, JEG3 cells lacked expression of classical progesterone receptors (PRs) but exhibited high expression of PGRMC1, a finding we confirmed in primary trophoblasts by mining sc-RNA seq data from human placenta. To investigate the role of PGRMC1 in progesterone signaling, we used CRISPR/Cas9 technology to knockout PGRMC1 in JEG3 cells. PGRMC1-deficient cells showed a diminished response to progesterone stimulation. Furthermore, we found that the progesterone antagonist RU486 inhibited ELF3 expression in a PGRMC1-dependent manner, suggesting that RU486 acts as a progesterone antagonist by competing for receptor binding. Additionally, we found that RU486 inhibited cell invasion, an important process for successful pregnancy, and this inhibitory effect was dependent on PGRMC1. Our findings highlight the crucial role of PGRMC1 in mediating the immunoregulatory effects of progesterone at the maternal-fetal interface.


Sujet(s)
Protéines membranaires , Progestérone , Récepteurs à la progestérone , Trophoblastes , Humains , Récepteurs à la progestérone/métabolisme , Femelle , Grossesse , Progestérone/métabolisme , Progestérone/pharmacologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Trophoblastes/métabolisme , Trophoblastes/immunologie , Placenta/immunologie , Placenta/métabolisme , Transduction du signal/immunologie , Échange foetomaternel/immunologie , Implantation embryonnaire/immunologie
12.
Immunol Rev ; 323(1): 288-302, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38445769

RÉSUMÉ

Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.


Sujet(s)
Exposition maternelle , Humains , Femelle , Grossesse , Animaux , Exposition maternelle/effets indésirables , Effets différés de l'exposition prénatale à des facteurs de risque/immunologie , Immunité acquise d'origine maternelle , Microbiote/immunologie , Système immunitaire/immunologie , Système immunitaire/croissance et développement , Échange foetomaternel/immunologie , Placenta/immunologie
13.
Science ; 381(6664): 1286, 2023 09 22.
Article de Anglais | MEDLINE | ID: mdl-37733842

RÉSUMÉ

Shifting pools of antigen can influence pregnancy-induced immune tolerance.


Sujet(s)
Chimérisme , Foetus , Tolérance immunitaire , Échange foetomaternel , Enfant , Femelle , Humains , Grossesse , Échange foetomaternel/immunologie , Antigènes/immunologie , Foetus/cytologie , Foetus/immunologie
14.
Science ; 381(6664): 1324-1330, 2023 09 22.
Article de Anglais | MEDLINE | ID: mdl-37733857

RÉSUMÉ

Pregnancy confers partner-specific protection against complications in future pregnancy that parallel persistence of fetal microchimeric cells (FMcs) in mothers after parturition. We show that preexisting FMcs become displaced by new FMcs during pregnancy and that FMc tonic stimulation is essential for expansion of protective fetal-specific forkhead box P3 (FOXP3)-positive regulatory T cells (Treg cells). Maternal microchimeric cells and accumulation of Treg cells with noninherited maternal antigen (NIMA) specificity are similarly overturned in daughters after pregnancy, highlighting a fixed microchimeric cell niche. Whereas NIMA-specific tolerance is functionally erased by pregnancy, partner-specific resiliency against pregnancy complications persists in mothers despite paternity changes in intervening pregnancy. Persistent fetal tolerance reflects FOXP3 expression plasticity, which allows mothers to more durably remember their babies, whereas daughters forget their mothers with new pregnancy-imprinted immunological memories.


Sujet(s)
Chimérisme , Foetus , Tolérance immunitaire , Mémoire immunologique , Échange foetomaternel , Grossesse , Animaux , Femelle , Souris , Grossesse/immunologie , Antigènes/immunologie , Plasticité cellulaire , Foetus/cytologie , Foetus/immunologie , Facteurs de transcription Forkhead/immunologie , Échange foetomaternel/immunologie , Souris de lignée C57BL , Lymphocytes T régulateurs/immunologie
15.
Nature ; 619(7970): 595-605, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37468587

RÉSUMÉ

Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.


Sujet(s)
Échange foetomaternel , Trophoblastes , Utérus , Femelle , Humains , Grossesse , Artères/physiologie , Caduques/vascularisation , Caduques/cytologie , Caduques/immunologie , Caduques/physiologie , Premier trimestre de grossesse/génétique , Premier trimestre de grossesse/métabolisme , Premier trimestre de grossesse/physiologie , Trophoblastes/cytologie , Trophoblastes/immunologie , Trophoblastes/physiologie , Utérus/vascularisation , Utérus/cytologie , Utérus/immunologie , Utérus/physiologie , Échange foetomaternel/génétique , Échange foetomaternel/immunologie , Échange foetomaternel/physiologie , Facteurs temps , Protéomique , Analyse de profil d'expression de gènes , Jeux de données comme sujet , Âge gestationnel
16.
J Clin Virol ; 165: 105495, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37295035

RÉSUMÉ

BACKGROUNDS: Due to immaturity of their immune system, passive maternal immunization is essential for newborns during their first months of life. Therefore, in the current context of intense circulation of SARS-CoV-2, identifying factors influencing the transfer ratio (TR) of neutralizing antibodies against SARS-CoV-2 (NAb) appears important. METHODS: Our study nested in the COVIPREG cohort (NCT04355234), included mothers who had a SARS-CoV-2 PCR positive during their pregnancy and their newborns. Maternal and neonatal NAb levels were measured with the automated iFlash system. RESULTS: For the 173 mother-infant pairs included in our study, the median gestational age (GA) at delivery was 39.4 weeks of gestation (WG), and 29.7 WG at maternal SARS-CoV-2 infection. Using a multivariate logistic model, having a NAb TR above 1 was positively associated with a longer delay from maternal positive SARS-CoV-2 PCR to delivery (aOR 1.09, 95% CI: 1.03 - 1.17) and with a later GA at delivery (aOR = 1.58, 95% CI: 1.09 - 2.52). It was negatively associated with being a male newborn (aOR 0.21, 95% CI: 0.07 - 0.59). In 3rd trimester SARS-CoV-2 infected mothers, NAb TR was inferior to VZV, toxoplasmosis, CMV, measle and rubella's TR. However, in 1st or 2nd trimester infected mothers, only measle TR was different from NAb TR. CONCLUSION: Male newborn of mothers infected by SARS-CoV-2 during their pregnancy appear to have less protection against SARS-CoV-2 in their first months of life than female newborns. Measle TR was superior to NAb TR even in case of 1st or 2nd trimester maternal SARS-CoV-2 infection. Future studies are needed to investigate possible differences in transmission of NAb following infection vs vaccination and its impact on TR.


Sujet(s)
Anticorps neutralisants , COVID-19 , Maladies néonatales , Transmission verticale de maladie infectieuse , Échange foetomaternel , Complications de la grossesse , SARS-CoV-2 , SARS-CoV-2/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Échange foetomaternel/immunologie , Âge gestationnel , Humains , Mâle , Femelle , COVID-19/sang , COVID-19/immunologie , COVID-19/prévention et contrôle , Accouchement (procédure) , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Grossesse , Nouveau-né , Caractères sexuels , Vaccins contre la COVID-19 , Vaccination , Complications de la grossesse/sang , Complications de la grossesse/immunologie , Maladies néonatales/immunologie , Maladies néonatales/prévention et contrôle , Transmission verticale de maladie infectieuse/prévention et contrôle , Paris , Adulte
17.
Proc Natl Acad Sci U S A ; 119(15): e2113310119, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35377817

RÉSUMÉ

Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic­polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood­brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte­endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.


Sujet(s)
Barrière hémato-encéphalique , Cyclooxygenase 2 , Encéphalite , Échange foetomaternel , Microglie , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Barrière hémato-encéphalique/malformations , Barrière hémato-encéphalique/physiopathologie , Célécoxib/pharmacologie , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Inhibiteurs de la cyclooxygénase 2/pharmacologie , Encéphalite/immunologie , Femelle , Délétion de gène , Échange foetomaternel/immunologie , Souris , Microglie/enzymologie , Poly I-C/immunologie , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/immunologie
19.
Placenta ; 119: 8-16, 2022 03 04.
Article de Anglais | MEDLINE | ID: mdl-35066308

RÉSUMÉ

INTRODUCTION: Placenta accreta spectrum (PAS) disorder is one of the major complications resulting in maternal death and serious adverse pregnancy outcomes. Uterine damage - principally that associated with cesarean section - is the leading risk factor for the development of PAS. However, the underlying pathogenesis of PAS related to uterine damage remains unclear. METHODS: For this study, we constructed a mouse PAS model using hysterotomy to simulate a cesarean section in humans. Pregnant mice were sacrificed on embryonic days 12.5 (E12.5) and E17.5. Trophoblast invasion and placental vascularization were analyzed using Hematoxylin-Eosin (H&E) staining and immunohistochemistry (IHC), and the proportions of immune cells at the maternal-fetal interface were analyzed using flow cytometry. We analyzed the expressions of genes in the decidua and placenta using RNA sequencing and subsequent validation by QPCR, and measured serum angiogenic factors by ELISA. RESULTS: Uterine damage led to increased trophoblast invasion and placental vascularization, with extensive changes to the immune-cell profiles at the maternal-fetal interface. The proportions of T and NK cells in the deciduas diminished significantly, with the decidual NK cells and M - 2 macrophages showing the greatest decline. The expression of TNF-α and IL4 was upregulated in the deciduas, while that of IFN-γ and IL10 was downregulated significantly. The expression of Mmp2, Mmp9, Mmp3, and Dock4 was significantly elevated in the placenta, and the serum levels of anti-angiogenic factors were significantly attenuated. DISCUSSION: Uterine damage can cause immune imbalance at the maternal-fetal interface, which may contribute to abnormal trophoblast invasion and enhanced vascularization of the mouse placenta.


Sujet(s)
Échange foetomaternel/immunologie , Néovascularisation physiologique , Procédures de chirurgie obstétrique/effets indésirables , Placenta accreta/étiologie , Trophoblastes/physiologie , Animaux , Femelle , Souris de lignée C57BL , Grossesse , Répartition aléatoire
20.
J Dev Orig Health Dis ; 13(1): 3-8, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-33526164

RÉSUMÉ

Little is known about the consequences of viral infection for pregnant woman or for the fetus. This issue became important with the appearance of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). The infection with SARS-CoV-2 causes a respiratory syndrome known as COVID-19. The fast spreading around the world and the fact that without a treatment or vaccine humans are completely exposed, converts emerging viral diseases in a significant risk for pregnant women and their infants. At this time, during SARS-CoV-2 pandemics pregnant women are not considered as a risk population and little is known about the effects of viral infections over the offspring although the amount of emerging evidence showing detrimental effects for the mother and the fetus. This issue highlights the importance to understand the effects of viral infections during pregnancy. In this work, we analyze the effects of viral infections, like SARS-CoV-2 and other related viruses during pregnancy over the mother and the consequences for the offspring.


Sujet(s)
COVID-19/complications , Complications infectieuses de la grossesse/virologie , SARS-CoV-2 , COVID-19/immunologie , Infections à coronavirus/complications , Femelle , Humains , Nouveau-né/immunologie , Transmission verticale de maladie infectieuse , Sous-type H1N1 du virus de la grippe A , Grippe humaine/complications , Échange foetomaternel/immunologie , Grossesse , Syndrome respiratoire aigu sévère/complications
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE