Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Hum Reprod ; 29(12)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38059603

RESUMEN

Growth-restricted placentae have a reduced vascular network, impairing exchange of nutrients and oxygen. However, little is known about the differentiation events and cell types that underpin normal/abnormal placental vascular formation and function. Here, we used 23-colour flow cytometry to characterize placental vascular/perivascular populations between first trimester and term, and in foetal growth restriction (FGR). First-trimester endothelial cells had an immature phenotype (CD144+/lowCD36-CD146low), while term endothelial cells expressed mature endothelial markers (CD36+CD146+). At term, a distinct population of CD31low endothelial cells co-expressed mesenchymal markers (CD90, CD26), indicating a capacity for endothelial to mesenchymal transition (EndMT). In FGR, compared with normal pregnancies, endothelial cells constituted 3-fold fewer villous core cells (P < 0.05), contributing to an increased perivascular: endothelial cell ratio (2.6-fold, P < 0.05). This suggests that abnormal EndMT may play a role in FGR. First-trimester endothelial cells underwent EndMT in culture, losing endothelial (CD31, CD34, CD144) and gaining mesenchymal (CD90, CD26) marker expression. Together this highlights how differences in villous core cell heterogeneity and phenotype may contribute to FGR pathophysiology across gestation.


Asunto(s)
Retardo del Crecimiento Fetal , Placenta , Humanos , Embarazo , Femenino , Placenta/metabolismo , Primer Trimestre del Embarazo , Retardo del Crecimiento Fetal/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Células Endoteliales/metabolismo
2.
Clin Sci (Lond) ; 137(5): 385-399, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36920079

RESUMEN

Human pregnancy is a highly orchestrated process requiring extensive cross-talk between the mother and the fetus. Extracellular vesicles released by the fetal tissue, particularly the placenta, are recognized as important mediators of this process. More recently, the importance of placental extracellular vesicle biodistribution studies in animal models has received increasing attention as identifying the organs to which extracellular vesicles are targeted to helps us understand more about this communication system. Placental extracellular vesicles are categorized based on their size into macro-, large-, and small-extracellular vesicles, and their biodistribution is dependent on the extracellular vesicle's particle size, the direction of blood flow, the recirculation of blood, as well as the retention capacity in organs. Macro-extracellular vesicles are exclusively localized to the lungs, while large- and small-extracellular vesicles show high levels of distribution to the lungs and liver, while there is inconsistency in the reporting of distribution to the spleen and kidneys. This inconsistency may be due to the differences in the methodologies employed between studies and their limitations. Future studies should incorporate analysis of placental extracellular vesicle biodistribution at the macroscopic level on whole animals and organs/tissues, as well as the microscopic cellular level.


Asunto(s)
Vesículas Extracelulares , Placenta , Animales , Embarazo , Femenino , Humanos , Placenta/metabolismo , Placentación , Distribución Tisular , Feto
3.
Mol Pharm ; 20(1): 23-40, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36332193

RESUMEN

Extracellular vesicles (EVs) are a group of cell-derived membrane vesicles of varying sizes that can be secreted by most cells. Depending on the type of cell they are derived from, EVs may contain a variety of cargo including proteins, lipids, miRNA, and DNA. Functionally, EVs play important roles in physiological and pathological processes through intercellular communication. While there has already been significant literature on the involvement of EVs in neurological and cardiovascular disease as well as cancer, recent evidence suggests that EVs may also play a role in mediating inflammatory eye diseases. This paper summarizes current advancements in ocular EV research as well as new ways by which EVs may be utilized as novel biomarkers of or therapeutics for inflammatory eye diseases.


Asunto(s)
Vesículas Extracelulares , Oftalmopatías , MicroARNs , Neoplasias , Humanos , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Biomarcadores/metabolismo , Oftalmopatías/tratamiento farmacológico , Neoplasias/metabolismo
4.
Mol Pharm ; 20(4): 1898-1913, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36919912

RESUMEN

Recently, extracellular vesicles (EVs) have garnered considerable interest as potential vehicles for drug delivery, including gene therapy. Although EVs from diverse sources have been investigated, current techniques used in the field for EV generation limit large-scale EV production. The placenta is essentially a tissue transplant and has unique properties that allow it to avoid the maternal immune system making it likely that placental EVs will not generate inflammatory responses and will avoid clearance by the immune system. We propose that placental EVs produced from explant cultures are an efficient method to produce considerable quantities of EVs that would be safe to administer, and we hypothesize that placental EVs can be loaded with large exogenous plasmids. To this end, we trialed three strategies to load plasmid DNA into placental EVs, including loading via electroporation of placental tissue prior to EV isolation and loading directly into placental EVs via electroporation or direct incubation of the EVs in plasmid solution. We report that the placenta releases vast quantities of EVs compared to placental cells in monolayer cultures. We show successful loading of plasmid DNA into both large- and small-EVs following both exogenous loading strategies with more plasmid encapsulated in large-EVs. Importantly, direct incubation did not alter EV size nor quantity. Further, we showed that the loading efficiency into EVs was dependent on the exogenous plasmid DNA dose and the DNA size. These results provide realistic estimates of plasmid loading capacity into placental EVs using current technologies and showcase the potential of placental EVs as DNA delivery vehicles.


Asunto(s)
Vesículas Extracelulares , Placenta , Embarazo , Femenino , Humanos , ADN , Sistemas de Liberación de Medicamentos , Plásmidos/genética
5.
Am J Obstet Gynecol ; 2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38158074

RESUMEN

BACKGROUND: Women with normotensive pregnancy are at a reduced risk of developing cardiovascular disease postpartum compared with those who experience hypertensive conditions during pregnancy. However, the underlying mechanisms remain poorly understood. During normotensive pregnancy, vast numbers of placental extracellular vesicles are released into the maternal circulation, which protect endothelial cells from activation and alter maternal vascular tone. We hypothesized that placental extracellular vesicles play a mechanistic role in lowering the risk of cardiovascular disease following normotensive pregnancy. OBJECTIVE: This study aimed to investigate the long-term effects of placental extracellular vesicles derived from normotensive term placentae on the cardiovascular system and explore the mechanisms underlying their biological effects. STUDY DESIGN: Spontaneously hypertensive rats were injected with placental extracellular vesicles from normotensive term pregnancies (2 mg/kg each time, n=8) or vehicle control (n=9) at 3 months of age. Blood pressure and cardiac function were regularly monitored from 3 months to 15 months of age. The response of mesenteric resistance arteries to vasoactive substances was investigated to evaluate vascular function. Cardiac remodeling, small artery remodeling, and renal function were investigated to comprehensively assess the impact of placental extracellular vesicles on cardiovascular and renal health. RESULTS: Compared with vehicle-treated control animals, rats treated with normotensive placental extracellular vesicles exhibited a significantly lower increase in blood pressure and improved cardiac function. Furthermore, the vasodilator response to the endothelium-dependent agonist acetylcholine was significantly enhanced in the normotensive placental extracellular vesicle-treated spontaneously hypertensive rats compared with the control. Moreover, treatment with placental extracellular vesicles reduced wall thickening of small renal vessels and attenuated renal fibrosis. CONCLUSION: Placental extracellular vesicles from normotensive term pregnancies have long-lasting protective effects reducing hypertension and mitigating cardiovascular damage in vivo.

6.
Cell Mol Life Sci ; 79(7): 384, 2022 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-35753002

RESUMEN

The use of in vitro tools to study trophoblast differentiation and function is essential to improve understanding of normal and abnormal placental development. The relative accessibility of human placentae enables the use of primary trophoblasts and placental explants in a range of in vitro systems. Recent advances in stem cell models, three-dimensional organoid cultures, and organ-on-a-chip systems have further shed light on the complex microenvironment and cell-cell crosstalk involved in placental development. However, understanding each model's strengths and limitations, and which in vivo aspects of human placentation in vitro data acquired does, or does not, accurately reflect, is key to interpret findings appropriately. To help researchers use and design anatomically accurate culture models, this review both outlines our current understanding of placental development, and critically considers the range of established and emerging culture models used to study this, with a focus on those derived from primary tissue.


Asunto(s)
Placenta , Placentación , Diferenciación Celular , Femenino , Humanos , Embarazo , Células Madre , Trofoblastos
7.
Anal Chem ; 94(37): 12907-12918, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36067379

RESUMEN

Machine learning has had a significant impact on the value of spectroscopic characterization tools, particularly in biomedical applications, due to its ability to detect latent patterns within complex spectral data. However, it often requires extensive data preprocessing, including baseline correction and denoising, which can lead to an unintentional bias during classification. To address this, we developed two deep learning methods capable of fully preprocessing raw Raman spectroscopy data without any human input. First, cascaded deep convolutional neural networks (CNN) based on either ResNet or U-Net architectures were trained on randomly generated spectra with augmented defects. Then, they were tested using simulated Raman spectra, surface-enhanced Raman spectroscopy (SERS) imaging of chemical species, low resolution Raman spectra of human bladder cancer tissue, and finally, classification of SERS spectra from human placental extracellular vesicles (EVs). Both approaches resulted in faster training and complete spectral preprocessing in a single step, with more speed, defect tolerance, and classification accuracy compared to conventional methods. These findings indicate that cascaded CNN preprocessing is ideal for biomedical Raman spectroscopy applications in which large numbers of heterogeneous spectra with diverse defects need to be automatically, rapidly, and reproducibly preprocessed.


Asunto(s)
Placenta , Espectrometría Raman , Diagnóstico por Imagen , Femenino , Humanos , Aprendizaje Automático , Redes Neurales de la Computación , Embarazo , Espectrometría Raman/métodos
8.
Reproduction ; 164(4): 143-154, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35938796

RESUMEN

In brief: Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) have shown promise as off-the-shelf therapeutics; however, producing them in sufficient quantities can be challenging. In this study, MSCs were isolated from preimplantation equine embryos and used to produce EVs in two commercially available bioreactor designs. Abstract: Mesenchymal stromal cells (MSC) have recently been explored for their potential use as therapeutics in human and veterinary medicine applications, such as the treatment of endometrial inflammation and infertility. Allogeneic MSC-derived extracellular vesicles (EVs) may also provide therapeutic benefits with advantage of being an 'off-the-shelf' solution, provided they can be produced in large enough quantities, without contamination from bovine EVs contained in fetal bovine serum that is a common component of cell culture media. Toward this aim, we demonstrated the successful isolation and characterization of equine MSCs from preimplantation embryos. We also demonstrate that many of these lines can be propagated long-term in culture while retaining their differentiation potential and conducted a head-to-head comparison of two bioreactor systems for scalable EV production including in serum-free conditions. Based on our findings, the CELLine AD 1000 flasks enabled higher cell density cultures and significantly more EV production than the FiberCell system or conventional culture flasks. These findings will enable future isolation of equine MSCs and the scalable culture of their EVs for a wide range of applications in this rapidly growing field.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Embrión de Mamíferos , Vesículas Extracelulares/metabolismo , Caballos , Humanos , Células Madre Mesenquimatosas/metabolismo
9.
Int J Mol Sci ; 23(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36361901

RESUMEN

Placental extracellular vesicles (EVs) have increasingly been recognized as a major mediator of feto-maternal communication. However, the cellular and molecular mechanisms of the uptake of placental EVs by recipient cells are still not well-understood. We previously reported that placental EVs target a limited number of organs in vivo. In the current study, we investigated the mechanisms underlying the uptake of placental EVs into target cells. Placental EVs were derived from explant cultures of normal or preeclamptic placentae. The mechanisms underlying the uptake of placental EVs were elucidated, using the phagocytosis or endocytosis inhibitor, trypsin-treatment or integrin-blocking peptides. The endothelial cell activation was studied using the monocyte adhesion assay after the preeclamptic EVs exposure, with and/or without treatment with the integrin blocking peptide, YIGSR. The cellular mechanism of the uptake of the placental EVs was time, concentration and energy-dependent and both the phagocytosis and endocytosis were involved in this process. Additionally, proteins on the surface of the placental EVs, including integrins, were involved in the EV uptake process. Furthermore, inhibiting the uptake of preeclamptic EVs with YIGSR, reduced the endothelial cell activation. The interaction between the placental EVs and the recipient cells is mediated by integrins, and the cellular uptake is mediated by a combination of both phagocytosis and endocytosis.


Asunto(s)
Vesículas Extracelulares , Preeclampsia , Enfermedades Vasculares , Humanos , Femenino , Embarazo , Placenta/metabolismo , Células Endoteliales/metabolismo , Preeclampsia/metabolismo , Vesículas Extracelulares/metabolismo , Enfermedades Vasculares/metabolismo , Integrinas/metabolismo
10.
Reproduction ; 162(4): 319-330, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34397395

RESUMEN

Placentae from pregnancies with foetal growth restriction (FGR) exhibit poor oxygen and nutrient exchange, in part due to impaired placental vascular development. Placental mesenchymal stromal cells (pMSCs) reside in a perivascular niche, where they may influence blood vessel formation/function. However, the role of pMSCs in vascular dysfunction in FGR is unclear. To elucidate the mechanisms by which pMSCs may impact placental vascularisation we compared the transcriptomes of human pMSCs isolated from FGR (<5th centile) (n = 7) and gestation-matched control placentae (n = 9) using Affymetrix microarrays. At the transcriptome level, there were no statistically significant differences between normal and FGR pMSCs; however, several genes linked to vascular function exhibited notable fold changes, and thus the dataset was used as a hypothesis-generating tool for possible dysfunction in FGR. Genes/proteins of interest were followed up by real-time PCR, western blot and immunohistochemistry. Gene expression of ADAMTS1 and FBLN2 (fibulin-2) were significantly upregulated, whilst HAS2 (hyaluronan synthase-2) was significantly downregulated, in pMSCs from FGR placentae (n = 8) relative to controls (n = 7, P < 0.05 for all). At the protein level, significant differences in the level of fibulin-2 and hyaluronan synthase-2, but not ADAMTS1, were confirmed between pMSCs from FGR and control pregnancies by Western blot. All three proteins demonstrated perivascular expression in third-trimester placentae. Fibulin-2 maintains vessel elasticity, and its increased expression in FGR pMSCs could help explain the increased distensibility of FGR blood vessels. ADAMTS1 and hyaluronan synthase-2 regulate angiogenesis, and their differential expression by FGR pMSCs may contribute to the impaired angiogenesis in these placentae.


Asunto(s)
Retardo del Crecimiento Fetal , Células Madre Mesenquimatosas , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Placenta/metabolismo , Embarazo
11.
Angiogenesis ; 23(2): 105-117, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31707538

RESUMEN

A branched vascular network is crucial to placental development and is dependent on factors such as vascular endothelial growth factor (VEGF), placental growth factor (PlGF), angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2), soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng) to regulate blood vessel growth. Imbalances in these factors can lead to aberrant placental vascular development. Throughout pregnancy, these factors are also released into the maternal circulation to aid in adapting the maternal cardiovascular system to pregnancy. Increased secretion of anti-angiogenic factors can lead to the development of an anti-angiogenic state in the mother and contribute to the development of pregnancy pathologies such as pre-eclampsia and foetal growth restriction (FGR). Thus, what are commonly referred to as 'angiogenic factors' have distinct functions in the maternal and placental circulations making this a misnomer. Indeed, technical issues in this field such as assay methodology and lack of data considering different placental cell types mean that the physiological roles of these factors in the maternal and placental circulations are frequently muddled in the literature. This review aims to (1) unpick the distinct roles of factors that influence placental vascular development and separate these from the roles of the same factors within the maternal circulation in normal pregnancy and (2) critically assess how imbalances may contribute to the distinct pathophysiological mechanisms underlying pregnancy disorders. Together, this critical assessment of the field endeavours to improve our ability to accurately use these factors as predictive/diagnostic biomarkers in the future.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/fisiología , Placenta/irrigación sanguínea , Circulación Placentaria , Complicaciones del Embarazo , Inhibidores de la Angiogénesis/farmacología , Angiopoyetinas/fisiología , Endoglina/fisiología , Femenino , Humanos , Placenta/efectos de los fármacos , Placenta/fisiología , Factor de Crecimiento Placentario/fisiología , Circulación Placentaria/efectos de los fármacos , Circulación Placentaria/genética , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
12.
J Autoimmun ; 111: 102437, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32224053

RESUMEN

Women with antiphospholipid antibodies (aPL) have increased risks of pregnancy complications, including a ten-fold increased risk of preeclampsia, which is potentially triggered by the release of placental toxins. Previously, aPL were shown to enter the outer layer of the placenta, the syncytiotrophoblast, associate with mitochondria, and alter mitochondrial function. We hypothesised that aPL may also increase mitochondrial reactive oxygen species (ROS) production, leading to cellular dysfunction and release of toxins. First trimester placental explants were incubated with monoclonal aPL, ID2 and IIC5 (25, 50, and 100 µg/mL), for 3 h at 37 °C and ROS production followed using CellROX Deep Red. In addition, the candidate treatment compounds chloroquine, melatonin, and Mito-Q were tested at therapeutic concentrations for their ability to prevent ROS production. Mitochondria isolated from term placentae were incubated with fluorescently-labelled ID2, IIC5, or control IgG antibodies (2.5, 5, 10, or 20 µg/mL) for 30 min, and mitochondria with bound antibodies were quantified using flow cytometry. In addition, respirometry coupled with fluorimetry was used to interrogate explant mitochondrial respiration and ROS production following incubation with 25, 50, or 100 µg/mL ID2, IIC5, or control IgG for 3 h at 37 °C. ID2 increased explant ROS production in a manner that was completely prevented by the endocytosis inhibitor chloroquine, and partially prevented by the antioxidants melatonin and Mito-Q. Both ID2 and IIC5 displayed a greater ability to bind isolated mitochondria than control antibodies, and increased ROS production attributable to the mitochondrial enzyme glycerol 3-phosphate dehydrogenase (mGPDH). Our evidence supports the hypothesis that aPL interact with syncytiotrophoblast mitochondria, likely via the binding of cardiolipin and ß2 glycoprotein I in mitochondrial membranes, and induce ROS production which contributes to overall oxidative stress and placental dysfunction.


Asunto(s)
Mitocondrias/metabolismo , Placenta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trofoblastos/metabolismo , Anticuerpos Antifosfolípidos , Respiración de la Célula , Células Cultivadas , Cloroquina/farmacología , Femenino , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Humanos , Inmunidad Humoral , Melatonina/farmacología , Embarazo , Primer Trimestre del Embarazo
13.
Reproduction ; 160(6): R119-R128, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33112772

RESUMEN

Trophoblasts are unique epithelial cells found only in the placenta. It has been possible to isolate and maintain human trophoblasts in in vitro culture for many decades. During this period there have been a vast array of media and supplements reported for trophoblast culture and often the reasons for using the media and specific supplements employed in any given laboratory have been lost in the 'mists of time'. After a gradual development over many years this field has recently changed, with the publication of several reports of the isolation, growth and differentiation of human trophoblast stem or stem-like cells. This advance was made largely because of a greater understanding of the molecular pathways that control human trophoblasts and availability of media supplements that can be used to manipulate those pathways. We have searched the literature and here summarise many of the different media and supplements and describe how and why they were developed and are used to culture human trophoblasts.


Asunto(s)
Diferenciación Celular , Medios de Cultivo/farmacología , Trofoblastos/citología , Células Cultivadas , Humanos , Trofoblastos/efectos de los fármacos
14.
J Autoimmun ; 98: 103-112, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30594350

RESUMEN

Women with antiphospholipid antibodies (aPL) are at high risk for pregnancy complications, such as preeclampsia. We previously demonstrated that aPL recognizing ß2GPI promote an extravillous trophoblast pro-inflammatory, anti-migratory and anti-angiogenic profile similar to that seen in preeclampsia. Since preeclampsia in the absence of aPL may have an underlying infectious element, women with aPL may be at increased risk for preeclampsia or other adverse outcomes if an infection is present. Our objective was to determine the impact the common bacterial component, muramyl dipeptide (MDP), has on trophoblast responses to aPL. Herein, we report that bacterial MDP amplifies trophoblast IL-1ß expression, processing, and secretion in the presence of aPL through activation of NOD2. In the absence of MDP, NOD2 also mediates anti- ß2GPI antibody-induced trophoblast IL-1ß and VEGF secretion. Additionally, we report a role for extravillous trophoblast vimentin as a novel danger signal that contributes to the aPL-induced trophoblast IL-1ß production. Together our data indicate that NOD2 mediates trophoblast inflammatory and angiogenic responses to aPL alone, and mediates trophoblast inflammation in the presence of bacterial MDP. These findings suggest that a bacterial infection at the maternal-fetal interface may exacerbate the impact aPL have on trophoblast inflammation and, thus, on pregnancy outcome.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/metabolismo , Anticuerpos Antifosfolípidos/metabolismo , Antígenos Bacterianos/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Trofoblastos/inmunología , Síndrome Antifosfolípido , Línea Celular , Femenino , Humanos , Inflamación , Interleucina-1beta/metabolismo , Neovascularización Patológica , Preeclampsia , Embarazo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Physiology (Bethesda) ; 32(3): 234-245, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28404739

RESUMEN

The utero-placental circulation links the maternal and fetal circulations during pregnancy, ensuring adequate gas and nutrient exchange, and consequently fetal growth. However, our understanding of this circulatory system remains incomplete. Here, we discuss how the utero-placental circulation is established, how it changes dynamically during pregnancy, and how this may impact on pregnancy success, highlighting how we may address knowledge gaps through advances in imaging and computational modeling approaches.


Asunto(s)
Circulación Placentaria , Embarazo , Animales , Femenino , Desarrollo Fetal , Humanos , Ciclo Menstrual , Placenta/irrigación sanguínea , Placenta/fisiología , Útero/irrigación sanguínea , Útero/fisiología
17.
Immunol Cell Biol ; 2018 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-29604098

RESUMEN

Extracellular vesicles (EVs) extruded by the human placenta are increasingly being recognized as an essential mode of feto-maternal communication. In the past two decades, there has been an explosion of research into the roles that placental EVs play in modulating the maternal immune and cardiovascular systems during healthy pregnancies, as well as how this communication is altered in obstetric diseases. This review aims to introduce readers to the processes of placental EV formation and the cargos they carry, and also to collate and summarize the published literature that investigates the immunological effects of placental EVs throughout human pregnancy.

18.
Int J Gynecol Cancer ; 28(3): 545-552, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29040188

RESUMEN

OBJECTIVE: Ovarian cancer is a common gynecological cancer, and parity is negatively associated with the incidence of this disease. This negative association is hypothesized to be due in part to shifting the balance of estrogen and progesterone toward more progesterone and reduced ovulation during pregnancy. However, studies suggested that parity is also associated with estrogen-independent gynecological cancers suggesting balance of hormones may not be the only protective factor. Extracellular vesicles (EVs) play an important role in cell-to-cell communication in physiological and pathological conditions. During pregnancy, large amounts of EVs are extruded from the placenta, and they seem to be involved in the remarkable adaptation of a woman's body to normal pregnancy. We hypothesized that EVs extruded from the placenta play a role in this protective effect. METHODS: Placental EVs were collected from first-trimester placentae, and cancer cell EVs were isolated from ovarian cancer cells. The EVs were exposed to ovarian cancer cells for 48 hours. The proliferation of cancer cells and the cell cycle were measured. In addition, phagocytosis of deported placental EVs by cancer cells was also measured. RESULTS: The proliferation of cancer cells was significantly reduced by treatment with placental EVs (P = 0.001, analysis of variance), but not EVs from monocytes (P = 0.195), compared with untreated cancer cells. Furthermore, placental EVs also prevented the proliferation of cancer cells induced by cancer cell-derived EVs (P = 0.001). This inhibition of proliferation of ovarian cancer cells was partially due to phagocytosis of placental EVs by cancer cells. Phagocytosis of placental EVs delayed progression through the cell cycle. Calreticulin, a phagocytic "eat me" signal carried by placental EVs significantly inhibited ovarian cancer growth (P = 0.001). CONCLUSIONS: Our data demonstrated that EVs extruded from the placenta prevented ovarian cancer cell growth by a mechanism that involved delaying progression of the cell cycle after phagocytosis of the EVs.


Asunto(s)
Vesículas Extracelulares/trasplante , Neoplasias Ováricas/terapia , Placenta/ultraestructura , Calreticulina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Vesículas Extracelulares/patología , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Neoplasias Ováricas/ultraestructura , Fagocitosis , Placenta/trasplante , Embarazo , Proteínas Recombinantes/farmacología , Células THP-1
19.
Reproduction ; 153(6): 835-845, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28356498

RESUMEN

Throughout human gestation, the placenta extrudes vast quantities of extracellular vesicles (EVs) of different sizes into the maternal circulation. Although multinucleated macro-vesicles are known to become trapped in the maternal lungs and do not enter the peripheral circulation, the maternal organs and cells that smaller placental micro-vesicles interact with in vivo remain unknown. This study aimed to characterise the interaction between placental micro-vesicles and endothelial cells in vitro and to elucidate which organs placental micro-vesicles localise to in vivo Placental macro- and micro-vesicles were isolated from cultured human first trimester placental explants by sequential centrifugation and exposed to human microvascular endothelial cells for up to 72 h. In vivo, placental macro- and micro-vesicles were administered to both non-pregnant and pregnant CD1 mice, and after two or 30 min or 24 h, organs were imaged on an IVIS Kinetic Imager. Placental EVs rapidly interacted with endothelial cells via phagocytic and clathrin-mediated endocytic processes in vitro, with over 60% of maximal interaction being achieved by 30 min of exposure. In vivo, placental macro-vesicles were localised exclusively to the lungs regardless of time of exposure, whereas micro-vesicles were localised to the lungs, liver and kidneys, with different distribution patterns depending on the length of exposure and whether the mouse was pregnant or not. The fact that placental EVs can rapidly interact with endothelial cells and localise to different organs in vivo supports that different size fractions of placental EVs are likely to have different downstream effects on foeto-maternal communication.


Asunto(s)
Células Endoteliales/fisiología , Vesículas Extracelulares/fisiología , Placenta/fisiología , Animales , Línea Celular , Femenino , Humanos , Ratones , Embarazo
20.
Mol Hum Reprod ; 22(7): 465-74, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27029214

RESUMEN

STUDY QUESTION: What is the role of microRNAs (miRs) in antiphospholipid antibody (aPL)-induced trophoblast inflammation? SUMMARY ANSWER: aPL-induced up-regulation of trophoblast miR-146a-3p is mediated by Toll-like receptor 4 (TLR4), and miR-146a-3p in turn drives the cells to secrete interleukin (IL)-8 by activating the RNA sensor, TLR8. WHAT IS KNOWN ALREADY: Obstetric antiphospholipid syndrome (APS) is an autoimmune disorder characterized by circulating aPL and an increased risk of pregnancy complications. We previously showed that aPL recognizing beta2 glycoprotein I (ß2GPI) elicit human first trimester trophoblast secretion of IL-8 by activating TLR4. Since some miRs control TLR responses, their regulation in trophoblast cells by aPL and functional role in the aPL-mediated inflammatory response was investigated. miRs can be released from cells via exosomes, and therefore, miR exosome expression was also examined. A panel of miRs was selected based on their involvement with TLR signaling: miR-9; miR-146a-5p and its isomiR, miR-146a-3p; miR-155, miR-210; and Let-7c. Since certain miRs can activate the RNA sensor, TLR8, this was also investigated. STUDY DESIGN, SIZE, DURATION: For in vitro studies, the human first trimester extravillous trophoblast cell line, HTR8 was studied. HTR8 cells transfected to express a TLR8 dominant negative (DN) were also used. Plasma was evaluated from pregnant women who have aPL, either with or without systemic lupus erythematous (SLE) (n = 39); SLE patients without aPL (n = 30); and healthy pregnant controls (n = 20). PARTICIPANTS/MATERIALS, SETTING, METHODS: Trophoblast HTR8 wildtype and TLR8-DN cells were incubated with or without aPL (mouse anti-human ß2GPI mAb) for 48-72 h. HTR8 cells were also treated with or without aPL in the presence and the absence of a TLR4 antagonist (lipopolysaccharide from Rhodobacter sphaeroides; LPS-RS), specific miR inhibitors or specific miR mimics. miR expression levels in trophoblast cells, trophoblast-derived exosomes and exosomes isolated from patient plasma were measured by qPCR. Trophoblast IL-8 secretion was measured by ELISA. MAIN RESULTS AND THE ROLE OF CHANCE: aPL significantly increased trophoblast cellular and exosome expression of miR-146a-5p, miR-146a-3p, miR-155 and miR-210. aPL-induced up-regulation of trophoblast miR-146a-5p, miR-146a-3p and miR-210, but not miR-155, was inhibited by the TLR4 antagonist, LPS-RS. While inhibition or overexpression of miR-146a-5p had no effect on aPL-induced trophoblast IL-8 secretion, miR-146a-3p inhibition significantly reduced this response. aPL-induced trophoblast IL-8 secretion was inhibited by the presence of the TLR8-DN. In the absence of aPL, transfection of trophoblast cells with a miR-146a-3p mimic significantly increased IL-8 secretion and this was inhibited by the presence of the TLR8-DN. Patients with aPL and adverse pregnancy outcomes (APOs) expressed significantly higher levels of circulating miR-146a-3p compared with healthy pregnant controls with no pregnancy complications (P < 0.05). LIMITATIONS, REASONS FOR CAUTION: While the enrichment of miR-146a-3p in trophoblast-derived exosomes support the role of this miR acting in a paracrine or endocrine manner through exosome delivery, this has not been demonstrated. However, miR-146a-3p may also exert its pro-inflammatory effect intracellularly within the same trophoblast cell targeted by aPL. WIDER IMPLICATIONS OF THE FINDINGS: These findings provide a novel mechanism of trophoblast inflammation through miRs activating RNA-sensing receptors. Furthermore, circulating exosomal-associated miR-146a-3p in APS patients may serve clinically as a biomarker for related APOs. STUDY FUNDING/COMPETING INTERESTS: This study was supported in part by grants from the American Heart Association (#10GRNT3640032 to V.M.A.), the March of Dimes Foundation (Gene Discovery and Translational Research Grant #6-FY12-255 to V.M.A.), NICHD, NIH (R01HD049446 to V.M.A.), the Gina M. Finzi Memorial Student Summer Fellowship from the Lupus Foundation of America (to S.M.G.), and the Yale University School of Medicine Medical Student Fellowship (to S.M.G.). The authors declare no competing financial interests. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Anticuerpos Antifosfolípidos/metabolismo , Interleucina-8/metabolismo , MicroARNs/metabolismo , Receptor Toll-Like 8/metabolismo , Trofoblastos/metabolismo , Síndrome Antifosfolípido/metabolismo , Femenino , Humanos , Embarazo , Primer Trimestre del Embarazo , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA