Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.653
Filtrar
1.
Am J Reprod Immunol ; 90(2): e13752, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37491922

RESUMO

PROBLEM: In the cell column of anchoring villi, the cytotrophoblast differentiates into extravillous trophoblast (EVT) and invades the endometrium in contact with maternal immune cells. Recently, chemokines were proposed to regulate the decidual immune response. To investigate the roles of chemokines around the anchoring villi, we examined the expression profiles of chemokines in the first-trimester trophoblast-derived Swan71 cells using a three-dimensional culture model. METHOD OF STUDY: The gene expressions in the spheroid-formed Swan71 cells were examined by microarray and qPCR analyses. The protein expressions were examined by immunochemical staining. The chemoattractant effects of spheroid-formed Swan71 cells were examined by migration assay using monocyte-derived THP-1 cells. RESULTS: The expressions of an EVT marker, laeverin, and matrix metalloproteases, MMP2 and MMP9, were increased in the spheroid-cultured Swan71 cells. Microarray and qPCR analysis revealed that mRNA expressions of various chemokines, CCL2, CCL7, CCL20, CXCL1, CXCL2, CXCL5, CXCL6, CXCL8, and CXCL10, in the spheroid-cultured Swan71 cells were up-regulated as compared with those in the monolayer-cultured Swan71 cells. These expressions were significantly suppressed by hypoxia. Migration assay showed that culture media derived from the spheroid-formed Swan71 cells promoted THP-1 cell migration. CONCLUSION: This study indicated that chemokine expressions in Swan71 cells increase under a spheroid-forming culture and the culture media have chemoattractant effects. Since three-dimensional cell assembling in the spheroid resembles the structure of the cell column, this study also suggests that chemokines play important roles in the interaction between EVT and immune cells in their early differentiation stage.


Assuntos
Trofoblastos , Humanos , Linhagem Celular , Quimiocinas/biossíntese , Trofoblastos/citologia , Trofoblastos/imunologia , Diferenciação Celular , Regulação da Expressão Gênica , RNA Mensageiro/genética , Movimento Celular , Oxigênio/metabolismo
2.
Nature ; 619(7970): 595-605, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37468587

RESUMO

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.


Assuntos
Troca Materno-Fetal , Trofoblastos , Útero , Feminino , Humanos , Gravidez , Artérias/fisiologia , Decídua/irrigação sanguínea , Decídua/citologia , Decídua/imunologia , Decídua/fisiologia , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/metabolismo , Primeiro Trimestre da Gravidez/fisiologia , Trofoblastos/citologia , Trofoblastos/imunologia , Trofoblastos/fisiologia , Útero/irrigação sanguínea , Útero/citologia , Útero/imunologia , Útero/fisiologia , Troca Materno-Fetal/genética , Troca Materno-Fetal/imunologia , Troca Materno-Fetal/fisiologia , Fatores de Tempo , Proteômica , Perfilação da Expressão Gênica , Conjuntos de Dados como Assunto , Idade Gestacional
3.
Biomolecules ; 12(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-35053216

RESUMO

A high number of leucocytes reside in the human endometrium and are distributed differentially during the menstrual cycle and pregnancy. During early pregnancy, decidual natural killer (dNK) cells are the most common type of natural killer (NK) cells in the uterus. The increase in the number of uterine NK (uNK) cells during the mid-secretory phase of the menstrual cycle, followed by further increase of dNK cells in early pregnancy, has heightened interest in their involvement during pregnancy. Extensive research has revealed various roles of dNK cells during pregnancy including the formation of new blood vessels, migration of trophoblasts, and immunological tolerance. The present review article is focused on the significance of NK cells during pregnancy and their role in pregnancy-related diseases. The article will provide an in-depth review of cellular and molecular interactions during pregnancy and related disorders, with NK cells playing a pivotal role. Moreover, this study will help researchers to understand the physiology of normal pregnancy and related complications with respect to NK cells, so that future research work can be designed to alleviate the complications.


Assuntos
Decídua/imunologia , Tolerância Imunológica , Células Matadoras Naturais/imunologia , Complicações na Gravidez/imunologia , Trofoblastos/imunologia , Feminino , Humanos , Gravidez
4.
Front Immunol ; 12: 743700, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858401

RESUMO

Pathological maternal inflammation and abnormal placentation contribute to several pregnancy-related disorders, including preterm birth, intrauterine growth restriction, and preeclampsia. TANK-binding kinase 1 (TBK1), a serine/threonine kinase, has been implicated in the regulation of various physiological processes, including innate immune response, autophagy, and cell growth. However, the relevance of TBK1 in the placental pro-inflammatory environment has not been investigated. In this study, we assessed the effect of TBK1 inhibition on lipopolysaccharide (LPS)-induced NLRP3 inflammasome activation and its underlying mechanisms in human trophoblast cell lines and mouse placenta. TBK1 phosphorylation was upregulated in the trophoblasts and placenta in response to LPS. Pharmacological and genetic inhibition of TBK1 in trophoblasts ameliorated LPS-induced NLRP3 inflammasome activation, placental inflammation, and subsequent interleukin (IL)-1 production. Moreover, maternal administration of amlexanox, a TBK1 inhibitor, reversed LPS-induced adverse pregnancy outcomes. Notably, TBK1 inhibition prevented LPS-induced NLRP3 inflammasome activation by targeting the mammalian target of rapamycin complex 1 (mTORC1). Thus, this study provides evidence for the biological significance of TBK1 in placental inflammation, suggesting that amlexanox may be a potential therapeutic candidate for treating inflammation-associated pregnancy-related complications.


Assuntos
Inflamassomos/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Complicações na Gravidez/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Trofoblastos/imunologia , Animais , Feminino , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Placenta/imunologia , Placenta/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Trofoblastos/metabolismo
5.
Front Immunol ; 12: 758281, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745133

RESUMO

The immune tolerance microenvironment is crucial for the establishment and maintenance of pregnancy at the maternal-fetal interface. The maternal-fetal interface is a complex system containing various cells, including lymphocytes, decidual stromal cells, and trophoblasts. Macrophages are the second-largest leukocytes at the maternal-fetal interface, which has been demonstrated to play essential roles in remodeling spiral arteries, maintaining maternal-fetal immune tolerance, and regulating trophoblast's biological behaviors. Many researchers, including us, have conducted a series of studies on the crosstalk between macrophages and trophoblasts at the maternal-fetal interface: on the one hand, macrophages can affect the invasion and migration of trophoblasts; on the other hand, trophoblasts can regulate macrophage polarization and influence the state of the maternal-fetal immune microenvironment. In this review, we systemically introduce the functions of macrophages and trophoblasts and the cell-cell interaction between them for the establishment and maintenance of pregnancy. Advances in this area will further accelerate the basic research and clinical translation of reproductive medicine.


Assuntos
Feto/imunologia , Tolerância Imunológica/imunologia , Macrófagos/fisiologia , Gravidez/imunologia , Trofoblastos/fisiologia , Animais , Comunicação Celular , Movimento Celular , Microambiente Celular , Citocinas/fisiologia , Vesículas Extracelulares/fisiologia , Feminino , Humanos , Ativação de Macrófagos , Macrófagos/classificação , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Trofoblastos/classificação , Trofoblastos/citologia , Trofoblastos/imunologia
6.
Front Immunol ; 12: 737401, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790194

RESUMO

Successful implantation requires the coordinated migration and invasion of trophoblast cells from out of the blastocyst and into the endometrium. This process relies on signals produced by cells in the maternal endometrium. However, the relative contribution of stroma cells remains unclear. The study of human implantation has major technical limitations, therefore the need of in vitro models to elucidate the molecular mechanisms. Using a recently described 3D in vitro models we evaluated the interaction between trophoblasts and human endometrial stroma cells (hESC), we assessed the process of trophoblast migration and invasion in the presence of stroma derived factors. We demonstrate that hESC promotes trophoblast invasion through the generation of an inflammatory environment modulated by TNF-α. We also show the role of stromal derived IL-17 as a promoter of trophoblast migration through the induction of essential genes that confer invasive capacity to cells of the trophectoderm. In conclusion, we describe the characterization of a cellular inflammatory network that may be important for blastocyst implantation. Our findings provide a new insight into the complexity of the implantation process and reveal the importance of inflammation for embryo implantation.


Assuntos
Movimento Celular , Implantação do Embrião , Endométrio/efeitos dos fármacos , Interleucina-17/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Trofoblastos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Adesão Celular , Diferenciação Celular , Linhagem Celular , Endométrio/imunologia , Endométrio/metabolismo , Feminino , Humanos , Interleucina-17/genética , Receptores Tipo I de Fatores de Necrose Tumoral/agonistas , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Via Secretória , Transdução de Sinais , Células Estromais/imunologia , Células Estromais/metabolismo , Trofoblastos/imunologia
7.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34785597

RESUMO

Zika virus (ZIKV) during pregnancy infects fetal trophoblasts and causes placental damage and birth defects including microcephaly. Little is known about the anti-ZIKV cellular immune response at the maternal-fetal interface. Decidual natural killer cells (dNK), which directly contact fetal trophoblasts, are the dominant maternal immune cells in the first-trimester placenta, when ZIKV infection is most hazardous. Although dNK express all the cytolytic molecules needed to kill, they usually do not kill infected fetal cells but promote placentation. Here, we show that dNK degranulate and kill ZIKV-infected placental trophoblasts. ZIKV infection of trophoblasts causes endoplasmic reticulum (ER) stress, which makes them dNK targets by down-regulating HLA-C/G, natural killer (NK) inhibitory receptor ligands that help maintain tolerance of the semiallogeneic fetus. ER stress also activates the NK activating receptor NKp46. ZIKV infection of Ifnar1 -/- pregnant mice results in high viral titers and severe intrauterine growth restriction, which are exacerbated by depletion of NK or CD8 T cells, indicating that killer lymphocytes, on balance, protect the fetus from ZIKV by eliminating infected cells and reducing the spread of infection.


Assuntos
Células Matadoras Naturais/imunologia , Trofoblastos/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Feto/imunologia , Antígenos HLA-C , Tolerância Imunológica , Camundongos , Placenta/imunologia , Placentação , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Receptores KIR
8.
Viruses ; 13(11)2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34835071

RESUMO

The mosquito-borne Rift Valley fever (RVF) is a prioritised disease that has been listed by the World Health Organization for urgent research and development of counteraction. Rift Valley fever virus (RVFV) can cause a cytopathogenic effect in the infected cell and induce hyperimmune responses that contribute to pathogenesis. In livestock, the consequences of RVFV infection vary from mild symptoms to abortion. In humans, 1-3% of patients with RVFV infection develop severe disease, manifested as, for example, haemorrhagic fever, encephalitis or blindness. RVFV infection has also been associated with miscarriage in humans. During pregnancy, there should be a balance between pro-inflammatory and anti-inflammatory mediators to create a protective environment for the placenta and foetus. Many viruses are capable of penetrating that protective environment and infecting the foetal-maternal unit, possibly via the trophoblasts in the placenta, with potentially severe consequences. Whether it is the viral infection per se, the immune response, or both that contribute to the pathogenesis of miscarriage remains unknown. To investigate how RVFV could contribute to pathogenesis during pregnancy, we infected two human trophoblast cell lines, A3 and Jar, representing normal and transformed human villous trophoblasts, respectively. They were infected with two RVFV variants (wild-type RVFV and RVFV with a deleted NSs protein), and the infection kinetics and 15 different cytokines were analysed. The trophoblast cell lines were infected by both RVFV variants and infection caused upregulation of messenger RNA (mRNA) expression for interferon (IFN) types I-III and inflammatory cytokines, combined with cell line-specific mRNA expression of transforming growth factor (TGF)-ß1 and interleukin (IL)-10. When comparing the two RVFV variants, we found that infection with RVFV lacking NSs function caused a hyper-IFN response and inflammatory response, while the wild-type RVFV suppressed the IFN I and inflammatory response. The induction of certain cytokines by RVFV infection could potentially lead to teratogenic effects that disrupt foetal and placental developmental pathways, leading to birth defects and other pregnancy complications, such as miscarriage.


Assuntos
Aborto Espontâneo/imunologia , Citocinas/imunologia , Vírus da Febre do Vale do Rift/patogenicidade , Trofoblastos/imunologia , Aborto Espontâneo/virologia , Morte Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Citocinas/genética , Feminino , Humanos , Inflamação , Mutação , Gravidez , RNA Mensageiro/genética , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Trofoblastos/virologia , Proteínas não Estruturais Virais/genética , Replicação Viral
9.
Reprod Biol Endocrinol ; 19(1): 150, 2021 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-34600537

RESUMO

The critical immune effectors, including T, B, and natural killer (NK) cells, dendritic cells, and macrophages participate in regulating immune responses during pregnancy. Among these immune cells, decidual NK (dNK) cells are involved in key placental development processes at the maternal-fetal interface, such as uterine spiral artery remodeling, trophoblast invasion, and decidualization. Mechanistically, dNK cells significantly influence pregnancy outcome by secreting cytokines, chemokines, and angiogenic mediators and by their interactions with trophoblasts and other decidual cells. MicroRNAs (miRNAs) are small non-coding RNA molecules that participate in the initiation and progression of human diseases. Although the functions of circulating miRNAs in pathological mechanism has been extensively studied, the regulatory roles of miRNAs in NK cells, especially in dNK cells, have been rarely reported. In this review, we analyze the effects of miRNA regulations of dNK cell functions on the immune system during gestation. We discuss aberrant expressions of certain miRNAs in dNK cells that may lead to pathological consequences, such as recurrent pregnancy loss (RPL). Interestingly, miRNA expression patterns are also different between dNK cells and peripheral NK (pNK) cells, and pNK cells in the first- and third-trimester of gestation. The dysregulation of miRNA plays a pivotal regulatory role in driving immune functions of dNK and pNK cells. Further understanding of the molecular mechanisms of miRNAs in dNK cells may provide new insights into the development of therapeutics to prevent pregnancy failure.


Assuntos
Decídua/imunologia , Células Matadoras Naturais/metabolismo , MicroRNAs/fisiologia , Gravidez , Aborto Habitual/genética , Aborto Habitual/imunologia , Aborto Habitual/patologia , Decídua/metabolismo , Decídua/patologia , Feminino , Humanos , MicroRNAs/metabolismo , Gravidez/genética , Gravidez/imunologia , Trofoblastos/imunologia , Trofoblastos/metabolismo
10.
J Immunol ; 207(10): 2433-2444, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34663619

RESUMO

Throughout gestation, the maternal immune system is tightly modulated to allow growth of a semiallogeneic fetus. During the third trimester, the maternal immune system shifts to a proinflammatory phenotype in preparation for labor. What induces this shift remains unclear. Cell-free fetal DNA (cffDNA) is shed by the placenta and enters maternal circulation throughout pregnancy. Levels of cffDNA are increased as gestation progresses and peak before labor, coinciding with a shift to proinflammatory maternal immunity. Furthermore, cffDNA is abnormally elevated in plasma from women with complications of pregnancy, including preterm labor. Given the changes in maternal immunity at the end of pregnancy and the role of sterile inflammation in the pathophysiology of spontaneous preterm birth, we hypothesized that cffDNA can act as a damage-associated molecular pattern inducing an inflammatory cytokine response that promotes hallmarks of parturition. To test this hypothesis, we stimulated human maternal leukocytes with cffDNA from primary term cytotrophoblasts or maternal plasma and observed significant IL-1ß and CXCL10 secretion, which coincides with phosphorylation of IFN regulatory factor 3 and caspase-1 cleavage. We then show that human maternal monocytes are crucial for the immune response to cffDNA and can activate bystander T cells to secrete proinflammatory IFN-γ and granzyme B. Lastly, we find that the monocyte response to cffDNA leads to vascular endothelium activation, induction of myometrial contractility, and PGE2 release in vitro. Our results suggest that the immune response to cffDNA can promote key features of the parturition cascade, which has physiologic consequences relevant to the timing of labor.


Assuntos
Ácidos Nucleicos Livres/imunologia , Feto/imunologia , Monócitos/imunologia , Parto/imunologia , Trofoblastos/imunologia , Feminino , Humanos , Gravidez
12.
Immunobiology ; 226(5): 152125, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34365089

RESUMO

NK cell development is affected by their cellular microenvironment and cytokines, including IL-15 and IL-18. NK cells can differentiate in secondary lymphoid organs, liver and within the uterus in close contact with trophoblast cells. The aim was to evaluate changes in the NK cell phenotype and function in the presence of IL-15, IL-18 and JEG-3, a trophoblast cell line. When cocultured with JEG-3 cells, IL-15 caused an increase in the number of NKG2D+ NK-92 cells and the intensity of CD127 expression. IL-18 stimulates an increase in the amount of NKp44+ NK-92 cells and in the intensity of NKp44 expression by pNK in the presence of trophoblast cells. NK-92 cell cytotoxic activity against JEG-3 cells increased only in presence of IL-18. Data on changes in the cytotoxic activity of NK-92 cells against JEG-3 cells in the presence of IL-15 and IL-18 indicate the modulation of NK cell function both by the cytokine microenvironment and directly by target cells. IL-15 and IL-18 were present in conditioned media (CM) from 1st and 3rd trimester placentas. In the presence of 1st trimester CM and JEG-3 cells, NK-92 cells showed an increase in the intensity of NKG2D expression. In the presence of 3rd trimester CM and JEG-3 cells, a decrease in the expression of NKG2D by NK-92 cells was observed. Thus, culturing of NK-92 cells with JEG-3 trophoblast cells stimulated a pronounced change in the NK cell phenotype, bringing it closer to the decidual NK cell-like phenotype.


Assuntos
Interleucina-15/imunologia , Interleucina-18/imunologia , Células Matadoras Naturais/imunologia , Trofoblastos/imunologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Feminino , Humanos , Fenótipo , Gravidez , Receptores de Células Matadoras Naturais/imunologia
13.
Front Immunol ; 12: 709466, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34367171

RESUMO

The placenta controls the growth of the fetus and ensures its immune protection. Key to these functions, the syncytiotrophoblast (SYN) is a syncytium formed by fusion of underlying mononuclear trophoblasts. The SYN covers the placental surface and is bathed in maternal blood to mediate nutritional and waste exchanges between the mother and fetus. The bacterial pathogen Listeria monocytogenes breaches the trophoblast barrier and infects the placental/fetal unit resulting in poor pregnancy outcomes. In this work, we analyzed the L. monocytogenes intracellular lifecycle in primary human trophoblasts. In accordance with previous studies, we found that the SYN is 20-fold more resistant to infection compared to mononuclear trophoblasts, forming a protective barrier to infection at the maternal interface. We show for the first time that this is due to a significant reduction in L. monocytogenes uptake by the SYN rather than inhibition of the bacterial intracellular division or motility. We here report the first transcriptomic analysis of L. monocytogenes-infected trophoblasts (RNA sequencing). Pathway analysis showed that infection upregulated TLR2, NOD-like, and cytosolic DNA sensing pathways, as well as downstream pro-inflammatory circuitry (NF-κB, AP-1, IRF4, IRF7) leading to the production of mediators known to elicit the recruitment and activation of maternal leukocytes (IL8, IL6, TNFα, MIP-1). Signature genes associated with poor pregnancy outcomes were also upregulated upon infection. Measuring the release of 54 inflammatory mediators confirmed the transcriptomic data and revealed sustained production of tolerogenic factors (IL-27, IL-10, IL-1RA, TSLP) despite infection. Both the SYN and mononuclear trophoblasts produced cytokines, but surprisingly, some cytokines were predominantly produced by the SYN (IL-8, IL-6) or by non-fused trophoblasts (TNFα). Collectively, our data support that trophoblasts act as placental gatekeepers that limit and detect L. monocytogenes infection resulting in a pro-inflammatory response, which may contribute to the poor pregnancy outcomes if the pathogen persists.


Assuntos
Inflamação/etiologia , Listeria monocytogenes/fisiologia , Trofoblastos/imunologia , Trofoblastos/microbiologia , Proteínas de Bactérias/fisiologia , Células Cultivadas , Quimiocinas/biossíntese , Citocinas/biossíntese , Feminino , Células Gigantes/imunologia , Humanos , Proteínas de Membrana/fisiologia , Gravidez , Resultado da Gravidez , Transcriptoma
14.
Front Immunol ; 12: 717884, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34381459

RESUMO

The syncytiotrophoblast (STB) of human placenta constitutively and throughout pregnancy produces and secretes exosomes - nanometer-sized membrane-bound extracellular vesicles from the endosomal compartment that convey cell-cell contact 'by proxy' transporting information between donor and recipient cells locally and at a distance. Released in the maternal blood, STB-derived exosomes build an exosomal gradient around the feto-placental unit acting as a shield that protects the fetus from maternal immune attack. They carry signal molecules and ligands that comprise distinct immunosuppressive protein signatures which interfere with maternal immune mechanisms, potentially dangerous for the ongoing pregnancy. We discuss three immunosuppressive signatures carried by STB exosomes and their role in three important immune mechanisms 1) NKG2D receptor-mediated cytotoxicity, 2) apoptosis of activated immune cells and 3) PD-1-mediated immunosuppression and priming of T regulatory cells. A schematic presentation is given on how these immunosuppressive protein signatures, delivered by STB exosomes, modulate the maternal immune system and contribute to the development of maternal-fetal tolerance.


Assuntos
Biomarcadores , Exossomos/metabolismo , Imunomodulação , Trofoblastos/imunologia , Trofoblastos/metabolismo , Apoptose/imunologia , Endossomos/imunologia , Endossomos/metabolismo , Exossomos/imunologia , Proteína Ligante Fas/metabolismo , Feminino , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/metabolismo , Tolerância Imunológica , Ligantes , Troca Materno-Fetal/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Gravidez , Proteoma , Proteômica/métodos
15.
FASEB J ; 35(8): e21754, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34191338

RESUMO

To obtain a successful pregnancy, the establishment of maternal-fetal tolerance and successful placentation are required to be established. Disruption of this immune balance and/or inadequate placental perfusion is believed to be associated with a lot of pregnancy-related complications, such as recurrent spontaneous abortion, pre-eclampsia, and fetal intrauterine growth restriction. Extravillous trophoblasts (EVTs) have the unique ability to instruct decidual immune cells (DICs) to develop a regulatory phenotype for fetal tolerance. Utilizing immortalized human first trimester extravillous trophoblast cells and primary EVTs, we found that DICs promote EVT function and placental development. We have previously shown that checkpoints T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are important for DIC function. In the present study, we showed that blockade of Tim-3 and CTLA-4 pathways leaded to the abnormal DICs-EVTs interaction, poor placental development, and increased fetal loss. Treatment with IL-4 and IL-10 could rescue the adverse effects of targeting Tim-3 and CTLA-4 on the pregnancy outcome. Hence, the reproductive safety must be a criterion considered in the assessment of immuno-therapeutic agents. In addition, IL-4 and IL-10 may represent novel therapeutic strategies to prevent pregnancy loss induced by checkpoint inhibition.


Assuntos
Antígeno CTLA-4/imunologia , Decídua/imunologia , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Trofoblastos/imunologia , Animais , Antígeno CTLA-4/antagonistas & inibidores , Comunicação Celular/imunologia , Células Cultivadas , Decídua/citologia , Perda do Embrião/imunologia , Feminino , Receptor Celular 2 do Vírus da Hepatite A/antagonistas & inibidores , Humanos , Tolerância Imunológica , Interleucina-10/administração & dosagem , Interleucina-4/administração & dosagem , Masculino , Troca Materno-Fetal/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Modelos Imunológicos , Placentação/imunologia , Gravidez , Resultado da Gravidez , Transdução de Sinais/imunologia , Trofoblastos/citologia
16.
J Reprod Immunol ; 146: 103347, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34175737

RESUMO

OBJECTIVES: Trophoblastic invasion at the maternal-fetal interface can affect pregnancy outcomes. To describe an intuitive theme trends and knowledge structure of trophoblastic invasion-related literature from a bibliometric perspective, and provide researchers with new research hotspots. STUDY DESIGN: The literature form PubMed database related to trophoblastic invasion from January 1, 2012 to April 30, 2021 were extracted, and then biclustering analysis, co-word analysis, strategy diagram and social network analysis were performed to provide immature, or newly emerging research hotspots for researchers. RESULTS: A total of 96 high-frequency medical subjects heading terms were extracted and classified into 6 clusters. Themes in the first and second quadrant of strategy diagram, including trophoblasts metabolism, placenta metabolism, pre-eclampsia, etc., as the mature parts of the research on trophoblastic invasion have been well developed. On the other hand, themes in the third and fourth quadrants of strategy diagram, such as embryo implantation and trophoblasts immunology, pregnancy complication, matrix metalloproteinase and trophoblasts metabolism, habitual abortion and trophoblasts metabolism, etc., are immature themes. Social network analysis suggests that themes at the edge, such as habitual abortion / metabolism, placenta / immunology, natural killer cells / physiology, natural killer cells / immunology, embryo implantation / immunology, are considered new research hotspots and have considerable research space. CONCLUSION: By analyzing the research hotspots related to trophoblastic invasion, immature themes and emerging hotspots deserve more attention and can be considered as hints when launching new research projects.


Assuntos
Bibliometria , Implantação do Embrião/imunologia , Pré-Eclâmpsia/imunologia , Trofoblastos/imunologia , Feminino , Humanos , Pré-Eclâmpsia/patologia , Gravidez , Trofoblastos/patologia
17.
Sci Rep ; 11(1): 12709, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34135407

RESUMO

Congenital toxoplasmosis is represented by the transplacental passage of Toxoplasma gondii from the mother to the fetus. Our studies demonstrated that T. gondii developed mechanisms to evade of the host immune response, such as cyclooxygenase (COX)-2 and prostaglandin E2 (PGE2) induction, and these mediators can be produced/stored in lipid droplets (LDs). The aim of this study was to evaluate the role of COX-2 and LDs during T. gondii infection in human trophoblast cells and villous explants. Our data demonstrated that COX-2 inhibitors decreased T. gondii replication in trophoblast cells and villous. In BeWo cells, the COX-2 inhibitors induced an increase of pro-inflammatory cytokines (IL-6 and MIF), and a decrease in anti-inflammatory cytokines (IL-4 and IL-10). In HTR-8/SVneo cells, the COX-2 inhibitors induced an increase of IL-6 and nitrite and decreased IL-4 and TGF-ß1. In villous explants, the COX-2 inhibitors increased MIF and decreased TNF-α and IL-10. Furthermore, T. gondii induced an increase in LDs in BeWo and HTR-8/SVneo, but COX-2 inhibitors reduced LDs in both cells type. We highlighted that COX-2 is a key factor to T. gondii proliferation in human trophoblast cells, since its inhibition induced a pro-inflammatory response capable of controlling parasitism and leading to a decrease in the availability of LDs, which are essentials for parasite growth.


Assuntos
Vilosidades Coriônicas/parasitologia , Ciclo-Oxigenase 2/metabolismo , Gotículas Lipídicas/metabolismo , Toxoplasma/crescimento & desenvolvimento , Trofoblastos/parasitologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Vilosidades Coriônicas/imunologia , Vilosidades Coriônicas/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Proteínas da Matriz Extracelular/metabolismo , Interações Hospedeiro-Parasita , Humanos , Interleucinas/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Nitritos/metabolismo , Toxoplasma/imunologia , Fator de Crescimento Transformador beta/metabolismo , Trofoblastos/imunologia , Trofoblastos/metabolismo
18.
J Reprod Immunol ; 146: 103331, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34030048

RESUMO

Human Leukocyte Antigen-G (HLA-G) prevents the activity of immune cells and is decreased in women with preeclampsia. We aimed to investigate the associations between circulating soluble HLA-G (sHLA-G) and 92 cardiovascular disease-related biomarkers from a previously published multiplex study in women with preeclampsia and controls. We found 15 markers significantly associated with circulating sHLA-G in univariate analyses. After multivariable adjusted regression, only proto-oncogene tyrosine-protein kinase Src (SRC) and vascular endothelial growth factor D were significantly associated with sHLA-G. Low SRC, previously observed in the circulation of preeclamptic women, may be regulated by low sHLA-G, and reflect decreased trophoblast differentiation and syncytical formation.


Assuntos
Antígenos HLA-G/sangue , Pré-Eclâmpsia/epidemiologia , Trofoblastos/imunologia , Biomarcadores/sangue , Estudos de Casos e Controles , Diferenciação Celular/imunologia , Cesárea , Feminino , Antígenos HLA-G/imunologia , Fatores de Risco de Doenças Cardíacas , Humanos , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/imunologia , Gravidez , Medição de Risco/métodos , Fator D de Crescimento do Endotélio Vascular/sangue , Quinases da Família src/sangue
19.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805739

RESUMO

As most recently demonstrated by the SARS-CoV-2 pandemic, congenital and perinatal infections are of significant concern to the pregnant population as compared to the general population. These outcomes can range from no apparent impact all the way to spontaneous abortion or fetal infection with long term developmental consequences. While some pathogens have developed mechanisms to cross the placenta and directly infect the fetus, other pathogens lead to an upregulation in maternal or placental inflammation that can indirectly cause harm. The placenta is a temporary, yet critical organ that serves multiple important functions during gestation including facilitation of fetal nutrition, oxygenation, and prevention of fetal infection in utero. Here, we review trophoblast cell immunology and the molecular mechanisms utilized to protect the fetus from infection. Lastly, we discuss consequences in the placenta when these protections fail and the histopathologic result following infection.


Assuntos
Imunidade , Placenta/imunologia , Placenta/virologia , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/virologia , Viroses/imunologia , Vírus/imunologia , Feminino , Feto/imunologia , Feto/virologia , Humanos , Placenta/patologia , Gravidez , Trofoblastos/imunologia , Trofoblastos/virologia
20.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809077

RESUMO

Pregnancy is a unique situation of physiological immunomodulation, as well as a strong Multiple Sclerosis (MS) disease modulator whose mechanisms are still unclear. Both maternal (decidua) and fetal (trophoblast) placental cells secrete extracellular vesicles (EVs), which are known to mediate cellular communication and modulate the maternal immune response. Their contribution to the MS disease course during pregnancy, however, is unexplored. Here, we provide a first phenotypic and functional characterization of EVs isolated from cultures of term placenta samples of women with MS, differentiating between decidua and trophoblast. In particular, we analyzed the expression profile of 37 surface proteins and tested the functional role of placental EVs on mono-cultures of CD14+ monocytes and co-cultures of CD4+ T and regulatory T (Treg) cells. Results indicated that placental EVs are enriched for surface markers typical of stem/progenitor cells, and that conditioning with EVs from samples of women with MS is associated to a moderate decrease in the expression of proinflammatory cytokines by activated monocytes and in the proliferation rate of activated T cells co-cultured with Tregs. Overall, our findings suggest an immunomodulatory potential of placental EVs from women with MS and set the stage for a promising research field aiming at elucidating their role in MS remission.


Assuntos
Vesículas Extracelulares/genética , Imunidade/genética , Esclerose Múltipla/genética , Proteoma/genética , Comunicação Celular/genética , Técnicas de Cocultura , Citocinas/genética , Decídua/imunologia , Decídua/metabolismo , Vesículas Extracelulares/imunologia , Feminino , Humanos , Imunomodulação/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Placenta/imunologia , Placenta/metabolismo , Gravidez , Linfócitos T Reguladores/imunologia , Trofoblastos/imunologia , Trofoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...