Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 14: 1060253, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37091848

RESUMEN

Uterine spiral artery remodeling is necessary for fetal growth and development as well as pregnancy outcomes. During remodeling, trophoblasts invade the arteries, replace the endothelium and disrupt the vascular smooth muscle, and are strictly regulated by the local microenvironment. Elevated glucose levels at the fetal-maternal interface are associated with disorganized placental villi and poor placental blood flow. Hyperglycemia disturbs trophoblast proliferation and invasion via inhibiting the epithelial-mesenchymal transition, altering the protein expression of related proteases (MMP9, MMP2, and uPA) and angiogenic factors (VEGF, PIGF). Besides, hyperglycemia influences the cellular crosstalk between immune cells, trophoblast, and vascular cells, leading to the failure of spiral artery remodeling. This review provides insight into molecular mechanisms and signaling pathways of hyperglycemia that influence trophoblast functions and uterine spiral artery remodeling.


Asunto(s)
Hiperglucemia , Trofoblastos , Embarazo , Femenino , Humanos , Trofoblastos/metabolismo , Placenta/fisiología , Factor de Crecimiento Placentario/metabolismo , Arterias , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo
2.
Immunology ; 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36562137

RESUMEN

Natural killer (NK) cells are known for their potent ability to kill stressed cells, whereas host cells infected with intra-cellular bacteria may also be benefit from the selective killing function of NK cells and survive. The mechanism of how NK cells protect host cells infected with intra-cellular bacteria is still unclear. Here, we discovered that decidual NK (dNK) cells cannot only eliminate intra-cellular bacteria which infected trophoblasts, but can also synthesize more lipids and transport lipids to trophoblasts to avoid their apoptosis. Mechanically, NK cells synthesize more lipids accompanied by increasing expression of apolipoprotein APOD. Lipids in NK cells can be delivered to trophoblast cells through APOD, maintaining adequate lipid droplet content and lipid metabolism homeostasis in trophoblasts. Blocking the APOD receptor LRP1 abolished lipid transport from NK cells to trophoblasts, and the reduction of lipid droplets caused by bacterial infection in trophoblast cells could not be restored, culminating in cell apoptosis. Our study provides new evidence for the immune surveillance and protective effect of NK cells on embryos during early pregnancy.

3.
Cell Mol Life Sci ; 79(12): 611, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36449080

RESUMEN

Deficiency of decidual NK (dNK) cell number and function has been widely regarded as an important cause of spontaneous abortion. However, the metabolic mechanism underlying the crosstalk between dNK cells and embryonic trophoblasts during early pregnancy remains largely unknown. Here, we observed that enriched glutamine and activated glutaminolysis in dNK cells contribute to trophoblast invasion and embryo growth by insulin-like growth factor-1 (IGF-1) and growth differentiation factor-15 (GDF-15) secretion. Mechanistically, these processes are dependent on the downregulation of EGLN1-HIF-1α mediated by α-ketoglutarate (α-KG). Blocking glutaminolysis with the GLS inhibitor BPTES or the glutamate dehydrogenase inhibitor EGCG leads to early embryo implantation failure, spontaneous abortion and/or fetal growth restriction in pregnant mice with impaired trophoblast invasion. Additionally, α-KG supplementation significantly alleviated pregnancy loss mediated by defective glutaminolysis in vivo, suggesting that inactivated glutamine/α-ketoglutarate metabolism in dNK cells impaired trophoblast invasion and induced pregnancy loss.


Asunto(s)
Aborto Espontáneo , Animales , Femenino , Ratones , Embarazo , Diferenciación Celular , Glutamina/farmacología , Factor 15 de Diferenciación de Crecimiento , Factor I del Crecimiento Similar a la Insulina , Ácidos Cetoglutáricos/farmacología
4.
Parasit Vectors ; 15(1): 337, 2022 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-36153598

RESUMEN

BACKGROUND: Infections are a major threat to human reproductive health because they can induce pregnancy failure, including recurrent abortion, stillbirth, and preterm birth. Toxoplasma gondii (T. gondii) infection can result in adverse pregnancy outcomes by affecting certain immune molecules and cytokines. However, the detailed mechanisms behind T. gondii-induced pregnancy failure are poorly understood. METHODS: Toxoplasma gondii-infected wild-type (WT) pregnant mice and 2B4 knockout (2B4-/-) pregnant mice were established for in vivo study. Human decidual natural killer (dNK) cells were cultured for in vitro study. Abnormal pregnancy outcomes were observed, and the expression of 2B4, functional molecules (CD69, CD107a, tumor necrosis factor alpha [TNF-α], interferon gamma [IFN-γ]), and signaling molecules (SHP-2, Fyn, p-ERK, p-P38) in dNK cells were detected by flow cytometry, Western blot, reverse transcriptase polymerase chain reaction (RT-PCR), and/or immunofluorescence. The direct interactions (2B4 interacts with SHP-2 and Fyn; SHP-2 interacts with p-P38 and 2B4; Fyn interacts with p-ERK and 2B4) were verified by co-immunoprecipitation (co-IP) in NK-92 cells. RESULTS: Here, results showed that 2B4 was significantly downregulated after T. gondii infection. Subsequently, infected 2B4-/- pregnant mice displayed worse pregnancy outcomes compared with infected WT pregnant mice. Also, increased TNF-α and IFN-γ expression and elevated dNK cell cytotoxicity were found in 2B4-/- pregnant mice during T. gondii infection. In contrast, reduced TNF-α and IFN-γ expression and decreased human dNK cell activity were found following 2B4 activation during T. gondii infection. Interestingly, results showed that 2B4 binds to adaptor SHP-2 or Fyn, which then triggers different signaling pathways to regulate TNF-α and IFN-γ expression in dNK cells during T. gondii infection. Further, SHP-2 binds 2B4 and p-P38 directly after 2B4 activation, which generates an inhibitory signal for TNF-α and IFN-γ in NK-92 cells. In addition, Fyn can bind to 2B4 and p-ERK after activation of 2B4, thereby inhibiting TNF-α and IFN-γ expression in NK-92 cells following T. gondii infection. CONCLUSIONS: These data suggest that 2B4 may be a novel danger-signaling molecule that is implicated in pregnancy failure during T. gondii infection. Unraveling the mechanism by which 2B4 regulates dNK cell activity will provide novel insights to aid our understanding of T. gondii-induced adverse pregnancy outcomes.


Asunto(s)
Nacimiento Prematuro , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Toxoplasma , Toxoplasmosis , Animales , Citocinas/metabolismo , Femenino , Humanos , Interferón gamma , Células Asesinas Naturales/metabolismo , Ratones , Embarazo , Resultado del Embarazo , Nacimiento Prematuro/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Immunol Rev ; 308(1): 168-186, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35582842

RESUMEN

Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.


Asunto(s)
Decidua , Células Asesinas Naturales , Femenino , Humanos , Tolerancia Inmunológica , Macrófagos , Embarazo , Linfocitos T
6.
Front Immunol ; 13: 771732, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35359988

RESUMEN

Cellular metabolism plays an important role in regulating both human and murine NK cell functions. However, it remains unclear whether cellular metabolic process impacts on the function of decidual NK cells (dNK), essential tissue-resident immune cells maintaining the homeostasis of maternal-fetal interface. Remarkably, we found that glycolysis blockage enhances dNK VEGF-A production but restrains its proliferation. Furthermore, levels of IFN-γ and TNF-α secreted by dNK get decreased when glycolysis or oxidative phosphorylation (OXPHOS) is inhibited. Additionally, glycolysis, OXPHOS, and fatty acid oxidation disruption has little effects on the secretion and the CD107a-dependent degranulation of dNK. Mechanistically, we discovered that the mammalian target of rapamycin complex 1 (mTORC1) signaling inhibition leads to decreased glycolysis and OXPHOS in dNK. These limited metabolic processes are associated with attenuated dNK functions, which include restricted production of cytokines including IFN-γ and TNF-α, diminished CD107a-dependent degranulation, and restrained dNK proliferation. Finally, we reported that the protein levels of several glycolysis-associated enzymes are altered and the mTORC1 activity is significantly lower in the decidua of women with recurrent pregnancy loss (RPL) compared with normal pregnancy, which might give new insights about the pathogenesis of RPL. Collectively, our data demonstrate that glucose metabolism and mTORC1 signaling support dNK functions in early pregnancy.


Asunto(s)
Aborto Habitual , Factor de Necrosis Tumoral alfa , Aborto Habitual/metabolismo , Animales , Decidua , Femenino , Humanos , Células Asesinas Naturales , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Embarazo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
7.
J Reprod Immunol ; 150: 103475, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35074692

RESUMEN

Decidual natural killer cells (dNK) have been the focus of many studies because of their unique roles in both the anti-tumor immune response and healthy placental formation. Revealing the immunological mechanisms by which they interact with their target cells may lead to a better understanding of immune evasion of certain tumor cells, including abnormal cells of the different forms of gestational trophoblast disease and miscarriages of immunologic origin. Efforts to perform functional immunological studies on dNK cells have been limited by difficulty obtaining sufficent quantities of cells and sustaining the dNK phenotype. A novel protocol was developed to isolate and culture dNK cells from fresh, term placentas and complete hydatidiform moles.The placental samples were collected from healthy women undergoing scheduled elective cesarean delivery. The molar samples were collected after evacuation and curettage. Tissue samples were made into single cell suspensions using mechanical and enzymatic degradation, followed by fluorescence-activated cell sorting (FACS) using surface markers. The dNK cells were then expanded in cell culture. Their surface markers and cytotoxicity were reassessed by flow cytometry and functional assays. The protocol produces high quantities of enriched dNK cells which can be sustained in cell culture for at least a month, preserving their phenotype and funcionality for a week.


Asunto(s)
Mola Hidatiforme , Neoplasias Uterinas , Decidua , Femenino , Humanos , Mola Hidatiforme/metabolismo , Células Asesinas Naturales , Placenta , Embarazo , Neoplasias Uterinas/metabolismo
8.
J Reprod Immunol ; 150: 103477, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35051828

RESUMEN

The eutopic secretory phase endometrium in endometriosis overproduces and releases a soluble immunosuppressive CD200 molecule (CD200L) and is populated by stromal cells that contain a truncated CD200 (CD200S) that promotes a proinflammatory environment. The CD200S+ cell population persists when pregnancy occurs and are abundant in the early pregnancy decidua of women with missed abortion. In the present study, CD200S+, CD56+, and CD68+ cells were enumerated in formalin-fixed paraffin-embedded tissue sections from women with endometriosis and non-endometriosis controls. CD200S+ cells were more numerous than CD68+ macrophages and were similar in number and location to CD56bright endometrial NK cells. In some endometria, there was an additional population of CD200S- CD56+ NK cells. In ectopic endometrial peritoneal deposits and in ectopic myometrial deposits (adenomyosis), CD200S+ cells were less frequent, consistent with the known paucity of CD56+ NK cells in sites of ectopic deposits. CD200S+ cell frequency was greater in stroma surrounding the smaller ectopic cystic deposits. Dual immunofluorescent antibody staining confirmed CD200S+ cells were CD56+ NK cells. CD200S+ NK cell frequency may be greater in endometriosis patients' endometrium and may affect embryo survival in early pregnancy. In our opinion, regulation of alternative splicing that results in CD200S rather than CD200L may provide new diagnostic and therapeutic options for women with endometriosis.


Asunto(s)
Endometriosis , Endometrio , Femenino , Humanos , Células Asesinas Naturales , Macrófagos , Embarazo , Útero
9.
Int J Biol Sci ; 17(15): 4377-4395, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803505

RESUMEN

Extracellular vesicles derived from trophoblasts (T-EVs) play an important role in pregnancy, but the mechanism is not entirely clear. In this study, we found that HLA-E, which is mostly confined to the cytoplasm of trophoblast cells, was secreted by T-EVs. The level of HLA-E in T-EVs from unexplained recurrent spontaneous abortion (URSA) patients was lower than that in normal pregnancy (NP) and RSA patients who had an abnormal embryo karyotype (AK-RSA). T-EVs promoted secretion of IFN-γ and VEGFα by decidual NK (dNK) cells from URSA patients via HLA-E, VEGFα was necessary for angiogenesis and trophoblast growth, and IFN-γ inhibited Th17 induction. Glycolysis and oxidative phosphorylation (OxPhos) were involved in this process. Glycolysis but not OxPhos of dNK cells facilitated by T-EVs was dependent on mTORC1 activation. Inhibition of T-EV production in vivo increased the susceptibility of mice to embryo absorption, which was reversed by transferring exogenous T-EVs. T-EVs promoted secretion of IFN-γ and VEGFα by dNK cells to maintain pregnancy via Qa-1 in abortion-prone mouse models. This study reveals a new mechanism of pregnancy maintenance mediated by HLA-E via T-EVs.


Asunto(s)
Decidua/citología , Vesículas Extracelulares/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/fisiología , Trofoblastos/fisiología , Animales , Línea Celular , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Ratones Desnudos , Placenta/citología , Embarazo , Técnicas de Cultivo de Tejidos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Antígenos HLA-E
10.
Cancers (Basel) ; 13(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34638439

RESUMEN

Natural Killer (NK) cells have been found to be anergic, exhausted and pro-angiogenic in cancers. NK cell from healthy donors, exposed to TGFß, acquire the CD56brightCD9+CD49a+ decidual-like-phenotype, together with decreased levels of NKG2D activation marker, increased levels of TIM-3 exhaustion marker, similar to cancer-associated NK cells. Tissue inhibitors of metalloproteases (TIMPs) exert dual roles in cancer. The role of TIMPs in modulating immune cells is a very novel concept, and the present is the first report studying their ability to contrast TGFß action on NK cells. Here, we investigated the effects of TIMP1 and TIMP2 recombinant proteins in hindering decidual-like markers in NK cells, generated by polarizing cytolytic NK cells with TGFß. The effects of TIMP1 or TIMP2 on NK cell surface antigens were determined by multicolor flow cytometry. We found that TIMP1 and TIMP2 were effective in interfering with TGFß induced NK cell polarization towards a decidual-like-phenotype. TIMP1 and TIMP2 counteracted the effect of TGFß in increasing the percentage of CD56bright, CD16-, CD9+ and CD49a+, and restoring normal levels for TIMP 1 and 2 also inhibited decrease levels of the activation marker NKG2D induced by TGFß and decreased the TGFß upregulated exhaustion marker TIM-3. NK cell degranulation capabilities against K562 cells were also decreased by TGFß and not by TIMP1 or TIMP2. TIMP1 treatment could partially restore degranulation marker CD107a expression. Treatment with recombinant TIMP-1 or TIMP-2 showed a trend, although not statistically significant, to decrease CD49a+ and TIM-3+ expression and increase NKG2D in peripheral blood NK cells exposed to conditioned media from colon cancer cell lines. Our results suggest a potential role of TIMPs in controlling the tumor-associated cytokine TGFß-induced NK cell polarization. Given the heterogeneity of released factors within the TME, it is clear that TGFß stimulation represents a model to prove TIMP's new properties, but it cannot be envisaged as a soloist NK cell polarizing agent. Therefore, further studies from the scientific community will help defining TIMPs immunomodulatory activities of NK cells in cancer, and their possible future diagnostic-therapeutic roles.

11.
Reprod Biol Endocrinol ; 19(1): 150, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600537

RESUMEN

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.


Asunto(s)
Decidua/inmunología , Células Asesinas Naturales/metabolismo , MicroARNs/fisiología , Embarazo , Aborto Habitual/genética , Aborto Habitual/inmunología , Aborto Habitual/patología , Decidua/metabolismo , Decidua/patología , Femenino , Humanos , MicroARNs/metabolismo , Embarazo/genética , Embarazo/inmunología , Trofoblastos/inmunología , Trofoblastos/metabolismo
12.
Cells ; 10(7)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209508

RESUMEN

The pathogenesis of many serious diseases, including cancer, is closely related to disturbances in the angiogenesis process. Angiogenesis is essential for the progression of tumor growth and metastasis. The tumor microenvironment (TME) has immunosuppressive properties, which contribute to tumor expansion and angiogenesis. Similarly, the uterine microenvironment (UME) exerts a tolerogenic (immunosuppressive) and proangiogenic effect on its cells, promoting implantation and development of the embryo and placenta. In the TME and UME natural killer (NK) cells, which otherwise are capable of killing target cells autonomously, enter a state of reduced cytotoxicity or anergy. Both TME and UME are rich with factors (e.g., TGF-ß, glycodelin, hypoxia), which support a conversion of NK cells to the low/non-cytotoxic, proangiogenic CD56brightCD16low phenotype. It is plausible that the phenomenon of acquiring proangiogenic and low cytotoxic features by NK cells is not only limited to cancer but is a common feature of different angiogenesis-dependent diseases (ADDs). In this review, we will discuss the role of NK cells in angiogenesis disturbances associated with cancer and other selected ADDs. Expanding the knowledge of the mechanisms responsible for angiogenesis and its disorders contributes to a better understanding of ADDs and may have therapeutic implications.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neovascularización Patológica/inmunología , Humanos , Modelos Biológicos , Microambiente Tumoral
13.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32822574

RESUMEN

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Bacterias/inmunología , Movimiento Celular/inmunología , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Células HeLa , Humanos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/inmunología , Placenta/microbiología , Embarazo , Ratas , Células THP-1 , Trofoblastos/microbiología
14.
Front Immunol ; 11: 349, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32218780

RESUMEN

Earlier data suggest that progesterone-induced blocking factor (PIBF) is involved in implantation. The present study therefore aims to investigate the consequences of functional PIBF deficiency during the peri-implantation period. CD1 female mice were injected intraperitoneally with 2 µg anti-PIBF monoclonal antibody on days 1.5 and 4.5 of pregnancy. The number of implantation sites and resorption rates were recorded on day 10.5. PIBF+ decidual NK cells and B cells were detected by immunohistochemistry or immunofluorescence. Decidual and peripheral NK activity was assessed by flow cytometry. A prime PCR array was used for determining the differential expression of genes involved in lymphocyte activation and Th1 or Th2 differentiation in CD4+ and CD8+ spleen cells from pregnant anti-PIBF-treated and control mice. Anti-PIBF treatment in the peri-implantation period resulted in impaired implantation and increased resorption rates in later pregnancy. The number of PIBF+ decidual NK cells decreased, while both decidual and peripheral NK activity increased in the anti-PIBF-treated mice. B cells were absent from the resorbed deciduas of anti-PIBF-treated mice. The genes implicated in T cell activation were significantly downregulated in CD4+ and increased in CD8+ of the anti-PIBF-treated animals. The gene for IL-4 was significantly downregulated in CD4+ cells while that of IL-12A was upregulated in CD8+ cells of anti-PIBF-treated animals. These data suggest that the lack of PIBF results in an impaired T cell activation, together with Th1 differentiation and increased NK activity, resulting in implantation failure.


Asunto(s)
Implantación del Embrión , Proteínas Gestacionales/fisiología , Linfocitos T/inmunología , Animales , Linfocitos B/inmunología , Diferenciación Celular , Citotoxicidad Inmunológica , Decidua/inmunología , Femenino , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Masculino , Ratones , Embarazo
15.
Am J Reprod Immunol ; 81(5): e13104, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30803068

RESUMEN

PROBLEM: The progesterone-regulated genes, PIBF and Gal-1, are key players in the feto-maternal immunological interaction. This study aims to investigate the expression of PIBF and Gal-1 in WT and progesterone receptor KO models as well as subsequent effects of PIBF on decidualization of stromal cells. METHOD OF THE STUDY: PRAKO, PRBKO and PRKO BALB/c mice were used for assessing the role of PR isoforms in PIBF induction. PIBF- and Gal-1 mRNA expression in the uterus was tested by real-time PCR. The effect of PIBF on decidualization of endometrial stromal cells was verified by anti-desmin immunofluorescence. Immunohistochemistry was used for testing PIBF expression in the uterus. Gal-1, ERα and PR positive decidual NK cells were detected by immunofluorescence. RESULTS: PIBF mRNA was significantly increased in progesterone-treated WT mice, but not in PRKO and PRAKO mice. PIBF protein expression was reduced in the endometria of PRKO and PRAKO, but not in PRBKO mice. During a 6-day culture, PIBF induced decidual transformation of endometrial stromal cells. PIBF expression in the mouse uterus was highest during the implantation window, while Gal-1 mRNA expression continuously increased between day 2.5 and day 11.5 of gestation. Decidual NK cells express Gal-1 and ERα, but not PR at day 7.5 murine pregnancy. CONCLUSION: PIBF produced via engagement of PRA, is highly expressed in the endometrium during the implantation window, and plays a role in decidualization. The concerted action of PIBF and Gal-1 might contribute to the low cytotoxic activity of decidual NK cells.


Asunto(s)
Benzamidas/metabolismo , Decidua/fisiología , Endometrio/metabolismo , Intercambio Materno-Fetal/inmunología , Proteínas Gestacionales/metabolismo , Receptores de Progesterona/metabolismo , Células del Estroma/fisiología , Tirosina/análogos & derivados , Animales , Células Cultivadas , Implantación del Embrión , Endometrio/citología , Femenino , Regulación de la Expresión Génica , Tolerancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Animales , Embarazo , Proteínas Gestacionales/genética , Receptores de Progesterona/genética , Tirosina/genética , Tirosina/metabolismo
16.
Am J Obstet Gynecol ; 221(3): 183-193, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30802436

RESUMEN

The frequencies of preeclampsia, fetal growth restriction, fetal demise, and low birthweight are lower in subsequent pregnancies. Enhanced maternal cardiovascular adaptation, shorter first and second stages of labor, and more robust lactation also have been observed in subsequent as compared with first pregnancies. We sought to investigate the cellular and molecular bases for better outcomes in subsequent pregnancies. Based on the knowledge that specialized immune cells at the maternal-fetal interface, decidual natural killer cells, promote development of the placental bed and conversion of the spiral arteries by secreting a myriad of angiogenic and growth factors, we asked whether decidual natural killer cells differ in subsequent as compared with first pregnancies. This idea stemmed from recent studies suggesting that natural killer cells, although part of the innate immune system, possess some features of adaptive immunity, including a certain type of immune cell memory, termed trained immunity. We found that decidual natural killer cells from parous women "remember pregnancy" and differ from decidual natural killer cells of primigravidae. Compared with the decidual natural killer cells of first pregnancy, these cells, that we termed pregnancy-trained decidual natural killer cells, express greater levels of the natural killer receptors NKG2C and leukocyte immunoglobulin-like receptor B1, which interact with ligands expressed on invasive trophoblasts. Furthermore, they secrete greater levels of several growth factors, including vascular endothelial growth factor α as well as interferon-γ, augmenting remodeling of the placental bed. We propose that this pregnancy-trained memory dwells in the epigenome, where memory of stimuli is known to persist even when the stimulus is no longer present. This epigenetic memory apparently resides in endometrial natural killer cells between pregnancies. We suggest that this trained memory, which we coined pregnancy-trained decidual natural killer cells, may be the missing link in the immune basis for enhanced subsequent pregnancy. Epigenetic memory (chromatin modification) also may afford a global explanation for additional findings of enhanced maternal cardiovascular adaptation, shorter first and second stages of labor, and more robust lactation. Understanding the molecular and cellular bases of improved outcomes of subsequent pregnancy may lead to the development of treatment modalities designed for women at high risk for pregnancy disorders originating at the maternal-fetal interface.


Asunto(s)
Epigénesis Genética/fisiología , Células Asesinas Naturales/fisiología , Paridad/fisiología , Complicaciones del Embarazo/fisiopatología , Resultado del Embarazo , Adaptación Fisiológica/fisiología , Biomarcadores/metabolismo , Decidua/fisiología , Femenino , Humanos , Lactancia/fisiología , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/metabolismo
17.
Front Immunol ; 10: 2730, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921098

RESUMEN

To establish a healthy pregnancy, maternal immune cells must tolerate fetal allo-antigens and remain competent to respond to infections both systemically and in placental tissues. Extravillous trophoblasts (EVT) are the most invasive cells of extra-embryonic origin to invade uterine tissues and express polymorphic Human Leucocyte Antigen-C (HLA-C) of both maternal and paternal origin. Thus, HLA-C is a key molecule that can elicit allogeneic immune responses by maternal T and NK cells and for which maternal-fetal immune tolerance needs to be established. HLA-C is also the only classical MHC molecule expressed by EVT that can present a wide variety of peptides to maternal memory T cells and establish protective immunity. The expression of paternal HLA-C by EVT provides a target for maternal NK and T cells, whereas HLA-C expression levels may influence how this response is shaped. This dual function of HLA-C requires tight transcriptional regulation of its expression to balance induction of tolerance and immunity. Here, we critically review new insights into: (i) the mechanisms controlling expression of HLA-C by EVT, (ii) the mechanisms by which decidual NK cells, effector T cells and regulatory T cells recognize HLA-C allo-antigens, and (iii) immune recognition of pathogen derived antigens in context of HLA-C.


Asunto(s)
Antígenos HLA-C/inmunología , Tolerancia Inmunológica , Inmunidad , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Placenta/metabolismo , Alelos , Decidua/inmunología , Decidua/metabolismo , Femenino , Regulación de la Expresión Génica , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/genética , Humanos , Tolerancia Inmunológica/genética , Inmunidad/genética , Inmunomodulación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Embarazo , Regiones Promotoras Genéticas , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trofoblastos/metabolismo
18.
Immunity ; 47(6): 1100-1113.e6, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262349

RESUMEN

Natural killer (NK) cells are present in large populations at the maternal-fetal interface during early pregnancy. However, the role of NK cells in fetal growth is unclear. Here, we have identified a CD49a+Eomes+ subset of NK cells that secreted growth-promoting factors (GPFs), including pleiotrophin and osteoglycin, in both humans and mice. The crosstalk between HLA-G and ILT2 served as a stimulus for GPF-secreting function of this NK cell subset. Decreases in this GPF-secreting NK cell subset impaired fetal development, resulting in fetal growth restriction. The transcription factor Nfil3, but not T-bet, affected the function and the number of this decidual NK cell subset. Adoptive transfer of induced CD49a+Eomes+ NK cells reversed impaired fetal growth and rebuilt an appropriate local microenvironment. These findings reveal properties of NK cells in promoting fetal growth. In addition, this research proposes approaches for therapeutic administration of NK cells in order to reverse restricted nourishments within the uterine microenvironment during early pregnancy.


Asunto(s)
Aborto Habitual/inmunología , Traslado Adoptivo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Desarrollo Fetal/inmunología , Retardo del Crecimiento Fetal/prevención & control , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Asesinas Naturales/trasplante , Aborto Habitual/genética , Aborto Habitual/patología , Adulto , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Microambiente Celular , Citocinas/genética , Citocinas/inmunología , Decidua/inmunología , Decidua/patología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/inmunología , Retardo del Crecimiento Fetal/patología , Feto , Regulación del Desarrollo de la Expresión Génica , Antígenos HLA-G/genética , Antígenos HLA-G/inmunología , Humanos , Integrina alfa1/genética , Integrina alfa1/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/genética , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Ratones , Ratones Endogámicos C57BL , Embarazo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología
19.
Am J Reprod Immunol ; 78(6)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28975683

RESUMEN

PROBLEM: The mechanisms underlying the regulation of decidual natural killer cells (dNKs) at the maternal-fetal interface are unclear. METHOD OF STUDY: Primary trophoblasts (TROs), decidual stromal cells (DSCs), and dNKs were cocultured, and responses to LAIR-2 (LAIR-1 inhibitor) and P4H shRNA (collagen inhibitor) were studied. RESULTS: Coculture of dNKs with primary TROs/DSCs resulted in downregulation of Th1 cytokine production by dNKs. These effects were abrogated by LAIR-2 and P4H shRNA. LAIR-1 binds to SHP-1, which in turn binds to JAK1 and JAK2. Further, the phosphorylation of STAT1/STAT4 and the expression of the downstream transcription factors T-bet and Helios in dNKs were decreased by collagen treatment and primary TROs/DSCs coculture. CONCLUSION: The JAK-STAT pathway and its downstream transcription factors T-bet and Helios are involved in the regulation of dNK function by collagen/LAIR-1 interaction, and this signaling mechanism may contribute to the maintenance of immune tolerance at the maternal-fetal interface.


Asunto(s)
Colágeno/metabolismo , Decidua/patología , Células Asesinas Naturales/inmunología , Células del Estroma/fisiología , Trofoblastos/fisiología , Células Cultivadas , Técnicas de Cocultivo , Colágeno/genética , Citocinas/metabolismo , Femenino , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Tolerancia Inmunológica , Quinasas Janus/metabolismo , Activación de Linfocitos , Intercambio Materno-Fetal , Embarazo , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , ARN Interferente Pequeño/genética , Receptores Inmunológicos/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología
20.
Cell Immunol ; 317: 9-17, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28438315

RESUMEN

Our current aim was to investigate whether injection of TGF-ß1 played an important role in improving abnormal pregnancy outcomes with T. gondii infection and how the TGF-ß1 regulated. Results showed that TGF-ß1 exhibited improved pregnancy outcomes induced by T. gondii infection. dNK cytotoxicity was increased with T. gondii infection while decreased with TGF-ß1 treatment. dNK cytotoxicity related NKG2D/DAP10 expression, perforin, granzyme, IFN-γ and killer subsets were all increased with T. gondii infection while decreased after TGF-ß1 treatment. In addition, anti-TGF-ß1 antibodies could aggregate the cytotoxicity of dNK cells and the levels of molecules above. These results indicated that TGF-ß1 treatment could improve the abnormal pregnancy outcomes with T. gondii infection by decreasing the cytotoxicity of dNK cells mediated by NKG2D/DAP10 pathway and killer subset. These results suggested that TGF-ß1 might be a potential immunoprotective method for the treatment of abnormal pregnancy outcomes following T. gondii infection.


Asunto(s)
Decidua/inmunología , Células Asesinas Naturales/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Resultado del Embarazo , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Células Cultivadas , Citotoxicidad Inmunológica , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Embarazo , Receptores Inmunológicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA