1
|
Wang CY, Wang M, Zhao CY, Zhou Q, Zhang XY, Wang FX, Dong JM, Du CP, Zhang CL, Dang Y, Yang AJ, Dong JF, Li M. ADAMTS-13 Prevents VWF-Mediated Gastric Cancer Metastasis. Arterioscler Thromb Vasc Biol 2025. [PMID: 40336476 DOI: 10.1161/atvbaha.125.322553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Gastric cancer invades local tissue extensively and metastasizes through the circulation to remote organs. Patients with metastasized gastric cancer have poor clinical outcomes. The vasculature in the cancer niche is developed poorly, thus allowing cancer cells to be released into the circulation. However, it is poorly understood how cancer cells adhere to and transmigrate through the fully developed endothelium in remote organs and what key adhesive ligands are involved in the process. Here, we report results from a study designed to investigate the role of hyperadhesive VWF (von Willebrand factor) in promoting the pulmonary metastasis of gastric cancer. METHODS We used mouse models to investigate the roles of hyperadhesive VWF in the pulmonary metastasis of gastric cancer. The findings from these mouse models were validated through in vitro experiments that specifically examined how VWF promoted gastric cancer-derived extracellular vesicles to activate endothelial cells and analyzed established databases of patients with gastric cancer. RESULTS VWF in cancer-bearing mice became hyperadhesive and mediated the adhesion of gastric cancer-derived extracellular vesicles to the endothelium, where gastric cancer-derived extracellular vesicles caused endothelial permeability and promoted the transmigration of cancer cells to the interstitial tissue of the lungs. Reducing VWF adhesive activity by the metalloprotease ADAMTS-13 (A disintegrin and metalloprotease with thrombospondin type motifs, type 13) prevented the pulmonary metastasis of gastric cancer cells in mice. We further validated the findings in mice through targeted in vitro experiments and by associating VWF with the outcomes of patients with gastric cancer through established databases of patients with gastric cancer using bioinformatics tools. CONCLUSIONS We show how VWF becomes hyperadhesive to promote the pulmonary metastasis of gastric cancer through its interaction with gastric cancer-derived extracellular vesicles and that the hyperadhesive activity of VWF is reduced by ADAMTS-13 to prevent the metastasis.
Collapse
Affiliation(s)
- Chen-Yu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
- Experimental Teaching Center of Basic Medicine, School of Basic Medical Science, Lanzhou University, China. (M.W.)
| | - Chan-Yuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Xiao-Yu Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | | | - Jia-Ming Dong
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Cun-Pu Du
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Chen-Li Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Yun Dang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
- Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China (Y.D.)
| | - Ai-Jun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA (J.-f.D.)
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle (J.-f.D.)
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, China. (C.-y.W., M.W., C.-y.Z., Q.Z., X.-y.Z., J.-m.D., C.-p.D., C.-l.Z., Y.D., A.-j.Y., M.L.)
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, China. (M.L.)
| |
Collapse
|
2
|
Escudero C, Vatish M. Review: The potential role of placental extracellular vesicles in blood-brain barrier disruption and neuroinflammation in preeclampsia. Placenta 2025:S0143-4004(25)00104-3. [PMID: 40229181 DOI: 10.1016/j.placenta.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
Preeclampsia is a complex pregnancy disorder characterized by hypertension and multisystem organ damage, notably affecting the liver, kidneys, and brain. Eclampsia, a severe form of preeclampsia, is marked by the sudden onset of generalized tonic-clonic seizures. Brain complications, including eclampsia, are responsible for 60-70 % of preeclampsia-related maternal deaths, particularly in low-income regions. Despite the significant impact of brain complications in preeclampsia, their underlying pathophysiology remains unclear. Evidence suggests that brain edema in preeclampsia and eclampsia results from disruption of the blood-brain barrier (BBB). Although direct analysis of the BBB is challenging, in vitro studies indicate that plasma from women with preeclampsia can compromise the BBB, with the specific circulating factors involved still unidentified. Among the potential culprits, recent findings highlight placental-derived small extracellular vesicles (PDsEVs) as key players in BBB disruption observed in preeclampsia. This review examines the role of PDsEVs in the pathophysiology of brain edema associated with preeclampsia, emphasizing areas for future research, including neuroinflammation and neuron dysfunction. Additionally, we discuss the protective role of magnesium sulfate in these processes.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile; Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
3
|
Li Q, Zheng T, Chen J, Li B, Zhang Q, Yang S, Shao J, Guan W, Zhang S. Exploring melatonin's multifaceted role in female reproductive health: From follicular development to lactation and its therapeutic potential in obstetric syndromes. J Adv Res 2025; 70:223-242. [PMID: 38692429 PMCID: PMC11976432 DOI: 10.1016/j.jare.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Melatonin is mainly secreted by the pineal gland during darkness and regulates biological rhythms through its receptors in the suprachiasmatic nucleus of the hypothalamus. In addition, it also plays a role in the reproductive system by affecting the function of the hypothalamic-pituitary-gonadal axis, and by acting as a free radical scavenger thus contributing to the maintenance of the optimal physiological state of the gonads. Besides, melatonin can freely cross the placenta to influence fetal development. However, there is still a lack of overall understanding of the role of melatonin in the reproductive cycle of female mammals. AIM OF REVIEW Here we focus the role of melatonin in female reproduction from follicular development to delivery as well as the relationship between melatonin and lactation. We further summarize the potential role of melatonin in the treatment of preeclampsia, polycystic ovary syndrome, endometriosis, and ovarian aging. KEY SCIENTIFIC CONCEPTS OF REVIEW Understanding the physiological role of melatonin in female reproductive processes will contribute to the advancement of human fertility and reproductive medicine research.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
4
|
Lee SH. The role of extracellular vesicles in embryo development: implications for reproductive health and therapeutic potential. Reprod Fertil Dev 2025; 37:RD24151. [PMID: 40153376 DOI: 10.1071/rd24151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/11/2025] [Indexed: 03/30/2025] Open
Abstract
Extracellular vesicles (EVs) contain various biological molecules, such as proteins, lipids, and diverse nucleic acids, which alter various physiological and pathological processes in recipient cells. This review focuses on the current understanding of the biological characteristics of EVs on embryo development and their potential therapeutic value in treating reproductive disorders. EVs play a crucial role in early embryo development, from fertilization to the pre-implantation stage, gastrulation, cell differentiation, and organogenesis. During the pre-implantation period, EVs interact with maternal reproductive tissue and promote implantation receptivity. In gastrulation, EVs regulate cell differentiation, contributing to tissue formation and maintenance. Abnormal bioactive molecules in EVs are closely related to developmental disorders. Thus, EVs have the potential to serve as biomarkers. Moreover, EVs can serve as therapeutic agents, delivering genetic material for targeted tissue/organs. The findings of this review highlight the potential role of EVs in intercellular signaling during embryo development. This can help advance assisted reproductive technologies and therapies to overcome infertility issues and developmental disorders.
Collapse
Affiliation(s)
- Seok Hee Lee
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
5
|
Winter HE, Murrieta-Coxca JM, Álvarez D, Henao-Restrepo J, Fuentes-Zacarías P, Arcila-Barrera S, Steiniger F, Groten T, Markert UR, Morales-Prieto DM. Enhanced capture of preeclampsia-derived extracellular vesicles from maternal plasma by monocytes and T lymphocytes. J Reprod Immunol 2025; 167:104417. [PMID: 39709894 DOI: 10.1016/j.jri.2024.104417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Released from trophoblast and other fetal cells, placental extracellular vesicles (EVs) reach the maternal peripheral blood and modulate immune responses. Increased EVs in plasma of preeclampsia (PE) patients indicate their involvement in the etiology of this condition. This study addresses the uptake of plasma EVs by peripheral blood mononuclear cells (PBMCs) and explores the underlying internalization mechanisms. Plasma EVs were isolated from women with normotensive pregnancy (EVNP) and those with PE (EVPE), and characterized by cryo-transmission electron microscopy, nanoparticle tracking analysis, Western blotting, flow cytometry, and micro bicinchoninic acid assay (micro-BCA). To investigate whether the origin of PBMCs affects uptake, samples from males, pregnant women, and non-pregnant women were included. Primary PBMCs and macrophages derived from the human leukemia monocytic cell line THP-1 were incubated with PKH-stained EVs, and uptake was assessed by flow cytometry and confocal microscopy. Key molecules involved in monocyte differentiation and macrophage function were evaluated in EV-treated cells using LEGENDplex™ assay and real-time polymerase chain reaction (RT-PCR). Independent of the PBMC source, EVs were mostly captured by monocytes and in a lower proportion by T lymphocytes. Capture of EVPE was higher than of EVNP in primary T lymphocytes, monocytes, and THP-1-derived macrophages. After inhibition by Wortmannin and Cytochalasin D, EV internalization by THP-1-derived macrophages was significantly inhibited but not completely abolished. No defined polarization profile of treated THP-1-derived macrophages could be identified. These findings provide evidence of EV modifications in PE, which enhance their uptake by monocytes and other immune cells, mainly through phagocytosis and endocytosis.
Collapse
Affiliation(s)
- Hephzibah E Winter
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | | - Daniel Álvarez
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany; Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | | | | | - Sebastian Arcila-Barrera
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany; Grupo de Investigación en Hormonas, Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Sede Bogotá, Bogotá 111321, Colombia
| | - Frank Steiniger
- Centre for Electron Microscopy, Jena University Hospital, Jena, Germany
| | - Tanja Groten
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| | | |
Collapse
|
6
|
Sandoval H, Ibáñez B, Contreras M, Troncoso F, Castro FO, Caamaño D, Mendez L, Escudero-Guevara E, Nualart F, Mistry HD, Kurlak LO, Vatish M, Acurio J, Escudero C. Extracellular Vesicles From Preeclampsia Disrupt the Blood-Brain Barrier by Reducing CLDN5. Arterioscler Thromb Vasc Biol 2025; 45:298-311. [PMID: 39665142 DOI: 10.1161/atvbaha.124.321077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The physiopathology of life-threatening cerebrovascular complications in preeclampsia is unknown. We investigated whether disruption of the blood-brain barrier, generated using circulating small extracellular vesicles (sEVs) from women with preeclampsia or placentae cultured under hypoxic conditions, impairs the expression of tight junction proteins, such as CLDN5 (claudin-5), mediated by VEGF (vascular endothelial growth factor), and activation of KDR (VEGFR2 [VEGF receptor 2]). METHODS We perform a preclinical mechanistic study using sEVs isolated from plasma of pregnant women with normal pregnancy (sEVs-NP; n=9), sEVs isolated from plasma of women with preeclampsia (sEVs-PE; n=9), or sEVs isolated from placentas cultured in normoxia (sEVs-Nor; n=10) or sEVs isolated from placentas cultured in hypoxia (sEVs-Hyp; n=10). The integrity of the blood-brain barrier was evaluated using in vitro (human [hCMEC/D3] and mouse [BEND/3 (brain endothelial cell 3)] brain endothelial cell lines) and in vivo (nonpregnant C57BL/6J mice [4-5 months old; n=13] injected with sEVs-Hyp) models. RESULTS sEVs-PE and sEVs-Hyp reduced total and membrane-associated protein CLDN5 levels (P<0.05). These results were negated with sEVs-PE sonication. sEVs-Hyp injected into nonpregnant mice generated neurological deficits and blood-brain barrier disruption, specifically in the posterior area of the brain, associated with brain endothelial cell uptake of sEVs, sEVs-Hyp high extravasation, and reduction in CLDN5 levels in the brain cortex. Furthermore, sEVs-PE and sEVs-sHyp had higher VEGF levels than sEVs-NP and sEVs-Nor. Human brain endothelial cells exposed to sEVs-PE exhibited a reduction in the activation of KDR. Reduction in CLDN5 observed in cells treated with sEVs-Hyp was further enhanced in cells treated with KDR selective inhibitor. CONCLUSIONS sEVs-PE disrupts the blood-brain barrier, an effect replicated with sEVs-Hyp, and involves reduced CLDN5 and elevated VEGF contained within these vesicles. However, our results do not support the participation of KDR activation in the downregulation of CLDN5 observed with sEVs-Hyp. These findings will improve our understanding of the pathophysiology of cerebrovascular alterations in women with preeclampsia.
Collapse
Affiliation(s)
- Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
| | - Belén Ibáñez
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
| | - Moisés Contreras
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
| | - Fidel O Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan, Chile (F.O.C., D.C., L.M.)
| | - Diego Caamaño
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan, Chile (F.O.C., D.C., L.M.)
| | - Lidice Mendez
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan, Chile (F.O.C., D.C., L.M.)
| | - Estefanny Escudero-Guevara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
- PhD Program in Biomedical Sciences, Universidad de Talca, Chile (E.E.-G.)
| | - Francisco Nualart
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile (F.N.)
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepción, Chile (F.N.)
| | - Hiten D Mistry
- Division of Women and Children's Health, School of Life Course and Population Sciences, King's College London, United Kingdom (H.D.M.)
| | - Lesia O Kurlak
- Stroke Trials Unit, School of Medicine, Faculty of Medicine & Health Sciences, University of Nottingham, United Kingdom (L.O.K.)
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom (M.V., C.E.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.)
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom (M.V., C.E.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (C.E.)
| |
Collapse
|
7
|
Murugesan S, Saravanakumar L, Sadayappan S, Kannappan R, Sinkey RG, Tubinis MD, Tita AN, Jilling T, Berkowitz DE. Placental extracellular vesicles from women with severe preeclampsia alter calcium homeostasis in cardiomyocytes: an ex vivo study. Am J Physiol Cell Physiol 2025; 328:C128-C138. [PMID: 39652412 DOI: 10.1152/ajpcell.00409.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024]
Abstract
Women with severe preeclampsia (sPE) exhibit a heightened risk of postpartum cardiovascular disease compared with those with normotensive pregnancies (NTP). Although placental extracellular vesicles (EVs) play a crucial role in feto-maternal communication, their impact on cardiomyocytes, particularly in the context of sPE, remains unclear. This study investigated the effect of sPE-associated placental EVs (sPE-Plex EVs) on cardiomyocyte calcium dynamics. We hypothesized that sPE-Plex EV mediates cardiomyocyte dysfunction by disrupting calcium signaling. EVs were isolated from plasma and placental explant culture (Plex) using precipitation methods and confirmed as Plex EVs by placental alkaline phosphatase (PLAP) activity and electron microscopy. Moreover, confocal microscopy confirmed the uptake of plasma EVs in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and Plex EVs by human AC-16 cardiomyocyte (hAC-16CM) cells. hiPSC-CM cells treated with sPE-EVs and hAC-16CM cells treated with sPE-Plex EVs exhibited significantly lower levels of stromal interaction molecule 1 (STIM1) and phospholamban (PLN) proteins compared with those treated with normotensive controls EVs, as confirmed by Western blot analysis. Treatment with sPE-Plex EVs also resulted in the downregulation of STIM1 and PLN proteins in murine cardiomyocyte (mCM) cells compared with treatment with NTP-Plex EVs. Our findings suggest that both plasma EVs and Plex EVs from sPE may alter calcium signaling in cardiac cells by downregulating calcium sensor proteins (STIM1 and PLN). Therefore, plasma EVs and Plex EVs from sPE pregnancies have adverse effects by altering calcium dynamics in hiPSC-CM, hAC-16CM, and mCM compared with normotensive control and potential impairment of cardiomyocyte function ex vivo.NEW & NOTEWORTHY This study unveils a novel link between the placenta and PE-linked heart dysfunction. We isolated and characterized placental EVs from pregnancies with sPE and normotensive controls. These plasma sPE-EVs, and sPE-Plex EVs disrupt calcium signaling in heart cells, potentially via reduced STIM1 and PLN proteins. This suggests both plasma sPE-EVs and sPE-Plex EVs cargo drive these disruptive effects. Identifying these cargo molecules (miRNAs or proteins) holds promise for new PE therapies targeting cardiac dysfunction.
Collapse
Affiliation(s)
- Saravanakumar Murugesan
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Lakshmi Saravanakumar
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Sakthivel Sadayappan
- Department of Cellular and Molecular Medicine, Sarver Heart Center, University of Arizona College of Medicine, Tucson, Arizona, United States
| | - Ramaswamy Kannappan
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Rachel G Sinkey
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Michelle D Tubinis
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Alan N Tita
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Dan E Berkowitz
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
8
|
Ramos A, Youssef L, Molina P, Torramadé-Moix S, Martinez-Sanchez J, Moreno-Castaño AB, Blasco M, Guillén-Olmos E, De Moner B, Pino M, Tortajada M, Camacho M, Borrell M, Crovetto F, Ramirez-Bajo MJ, Ventura-Aguiar P, Banon-Maneus E, Rovira J, Escolar G, Carreras E, Gratacos E, Diaz-Ricart M, Crispi F, Palomo M. Circulating extracellular vesicles and neutrophil extracellular traps contribute to endothelial dysfunction in preeclampsia. Front Immunol 2024; 15:1488127. [PMID: 39735539 PMCID: PMC11671372 DOI: 10.3389/fimmu.2024.1488127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background Preeclampsia (PE) is a pregnancy complication characterized by hypertension, proteinuria, endothelial dysfunction, and complement dysregulation. Placenta-derived extracellular vesicles (EVs), necessary in maternal-fetal communication, might contribute to PE pathogenesis. Moreover, neutrophil extracellular traps (NETs) play a pathogenic role in other complement-mediated pathologies, and their contribution in PE remains unexplored. Materials and methods EVs were isolated from PE (peEVs) and normotensive pregnant women sera. NETs were obtained incubating donor-pre-activated neutrophils with PE or control sera. Microvascular (HMEC) endothelial cells (ECs) were incubated with PE or control sera with or without (depleted sera) EVs or NETs, to assess changes in VCAM-1, ICAM-1, VE-cadherin, eNOS, VWF, ROS, and C5b-9 deposits. Results were expressed as fold increase vs. control. Results VWF, VCAM-1, and ROS expression was significantly higher in cells exposed to PE sera vs. control (12.3 ± 8.1, 3.6 ± 2.3, and 1.8 ± 0.2, respectively, p < 0.05), though significantly lower in cells exposed to depleted PE (dPE) sera (6.1 ± 2.7, 0.7 ± 0.6, and 1.2 ± 0.1, respectively, vs. control, p < 0.05). EC exposure to depleted control sera supplemented with peEVs (dC+peEVs) significantly increased VWF, VCAM-1, and ROS compared to non-supplemented sera (4.5 ± 0.3, 2.8 ± 2.0, and 1.4 ± 0.2, respectively, p < 0.05). ICAM-1, VE-cadherin, and C5b-9 did not differ among groups. ECs incubated with PE-NETs increased VWF and VCAM-1 and decreased VE-cadherin expression vs. control (4 ± 1.6, 5.9 ± 1.2, and 0.5 ± 0.1, respectively, p < 0.05), and notably increased C5b-9 deposit (7.5 ± 2.9, p < 0.05). ICAM-1 and ROS did not differ. Conclusions Both circulating EVs and NETs from PE pregnant women exhibit a deleterious effect on ECs. Whereas EVs trigger a pro-oxidant and proinflammatory state, NETs potentiate the activation of the complement system, as already described in PE.
Collapse
Affiliation(s)
- Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Patricia Molina
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Torramadé-Moix
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Miquel Blasco
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud RICORS2040, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Guillén-Olmos
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Marc Pino
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Marta Tortajada
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Marta Camacho
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Borrell
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Jose Ramirez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Ventura-Aguiar
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elisenda Banon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Eduard Gratacos
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Palomo
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematology External Quality Assessment Laboratory, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Popova AK, Vashukova ES, Illarionov RA, Maltseva AR, Pachuliia OV, Postnikova TB, Glotov AS. Extracellular Vesicles as Biomarkers of Pregnancy Complications. Int J Mol Sci 2024; 25:11944. [PMID: 39596014 PMCID: PMC11594130 DOI: 10.3390/ijms252211944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Extracellular vesicles (EVs) are double-membrane vesicles that facilitate intercellular communication and play a pivotal role in both physiological and pathological processes. A substantial body of evidence suggests that EVs play a role in the pathogenesis of various pregnancy complications. Because EVs can be detected in the peripheral blood, they are potential biomarkers for the early diagnosis of pregnancy complications and foetal developmental disorders. The majority of studies have demonstrated a correlation between alterations in the concentration of EVs and changes in their contents and the occurrence of pregnancy complications. Despite the current limitations in establishing a clear link between these findings and the pathogenesis of the disease, as well as the lack of sufficient evidence to support their use in clinical practice, it is noteworthy to highlight the potential role of specific miRNAs carried by EVs in the development of pregnancy complications. These include miR-210 and miR-136-5p for pre-eclampsia and gestational diabetes mellitus, miR-155, miR-26b-5p, miR-181a-5p, miR-495 and miR-374c for pre-eclampsia and preterm birth. The following miRNAs have been identified as potential biomarkers for preterm birth and gestational diabetes mellitus: miR-197-3p and miR-520h, miR-1323, miR-342-3p, miR-132-3p, miR-182-3p, miR-517-3p, miR-222-3p, miR-16-5p and miR-126-3p. Additionally, miR-127-3p has been linked to foetal growth restriction and preterm birth. Nevertheless, it would be premature to propose that EVs can be employed as biomarkers for pregnancy complications. Further research and the accumulation of results obtained using the methods proposed in the MISEV2023 guidelines will enable a definitive conclusion to be reached.
Collapse
Affiliation(s)
- Anastasiia K. Popova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Elena S. Vashukova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Roman A. Illarionov
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Anastasia R. Maltseva
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Olga V. Pachuliia
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Tatiana B. Postnikova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Andrey S. Glotov
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg 199034, Russia
| |
Collapse
|
10
|
Jeyarajah MJ, Patterson VS, Jaju Bhattad G, Zhao L, Whitehead SN, Renaud SJ. Placental extracellular vesicles promote cardiomyocyte maturation and fetal heart development. Commun Biol 2024; 7:1254. [PMID: 39363116 PMCID: PMC11450004 DOI: 10.1038/s42003-024-06938-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Congenital heart defects are leading causes of neonatal mortality and are often associated with placental abnormalities, but mechanisms linking placenta and heart development are poorly understood. Herein, we investigated a potential signaling network connecting the placenta and nascent heart in mice. We found that fetal hearts exposed to media conditioned by placental tissue or differentiated wild-type trophoblast stem (TS) cells, but not undifferentiated TS cells, showed increased heart rate and epicardial cell outgrowth. This effect was not observed when hearts were exposed to media from TS cells lacking OVO-Like 2, a transcription factor required for trophoblast differentiation and placental development. Trophoblasts released abundant extracellular vesicles into media, and these vesicles were sufficient to mediate cardio-promoting effects. Our findings provide a potential mechanism whereby the placenta communicates with the fetal heart to promote cardiac morphogenesis, and offers insight into the link between poor placentation and a higher incidence of heart defects.
Collapse
Affiliation(s)
- Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Violet S Patterson
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Lin Zhao
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
- Children's Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
11
|
Lee JC, Ray RM, Scott TA. Prospects and challenges of tissue-derived extracellular vesicles. Mol Ther 2024; 32:2950-2978. [PMID: 38910325 PMCID: PMC11403234 DOI: 10.1016/j.ymthe.2024.06.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Extracellular vesicles (EVs) are considered a vital component of cell-to-cell communication and represent a new frontier in diagnostics and a means to identify pathways for therapeutic intervention. Recently, studies have revealed the importance of tissue-derived EVs (Ti-EVs), which are EVs present in the interstitial spaces between cells, as they better represent the underlying physiology of complex, multicellular tissue microenvironments in biology and disease. EVs are native, lipid bilayer membraned nano-sized particles produced by all cells that are packaged with varied functional biomolecules including proteins, lipids, and nucleic acids. They are implicated in short- and long-range cellular communication and may elicit functional responses in recipient cells. To date, studies have often utilized cultured cells or biological fluids as a source for EVs that do not capture local molecular signatures of the tissue microenvironment. Recent work utilizing Ti-EVs has elucidated novel biomarkers for disease and provided insights into disease mechanisms that may lead to the development of novel therapeutic agents. Still, there are considerable challenges facing current studies. This review explores the vast potential and unique challenges for Ti-EV research and provides considerations for future studies that seek to advance this exciting field.
Collapse
Affiliation(s)
- Justin C Lee
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Roslyn M Ray
- Gene Therapy Research, CSL Behring, Pasadena, CA 91106, USA
| | - Tristan A Scott
- Center for Gene Therapy, City of Hope, Beckman Research Institute and Hematological Malignancy and Stem Cell Transplantation Institute, Duarte, CA 91010, USA.
| |
Collapse
|
12
|
Parvin A, Erabi G, Mohammadpour D, Maleki-Kakelar H, Sadeghpour S, Pashaei MR, Taheri-Anganeh M, Ghasemnejad-Berenji H. Infertility: Focus on the therapeutic potential of extracellular vesicles. Reprod Biol 2024; 24:100925. [PMID: 39018753 DOI: 10.1016/j.repbio.2024.100925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Infertility is a well-known problem that arises from a variety of reproductive diseases. Until now, researchers have tried various methods to restore fertility, including medication specific to the cause, hormone treatments, surgical removals, and assisted reproductive technologies. While these methods do produce results, they do not consistently lead to fertility restoration in every instance. The use of exosome therapy has significant potential in treating infertility in patients. This is because exosomes, microvesicles, and apoptotic bodies, which are different types of vesicles, play a crucial role in transferring bioactive molecules that aid in cell-to-cell communication. Reproductive fluids can transport a variety of molecular cargos, such as miRNAs, mRNAs, proteins, lipids, and DNA molecules. The percentage of these cargos in the fluids can be linked to their physiological and pathological status. EVs are involved in several physiological and pathological processes and offer interesting non-cellular therapeutic possibilities to treat infertility. EVs (extracellular vesicles) transplantation has been shown in many studies to be a key part of regenerating different parts of the reproductive system, including the production of oocytes and the start of sperm production. Nevertheless, the existing evidence necessitates testifying to the effectiveness of injecting EVs in resolving reproductive problems among humans. This review focuses on the current literature about infertility issues in both females and males, specifically examining the potential treatments involving extracellular vesicles (EVs).
Collapse
Affiliation(s)
- Ali Parvin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Donna Mohammadpour
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Obstetrics & Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
13
|
Weiss L, Uhrig W, Kelliher S, Szklanna PB, Prendiville T, Comer SP, Edebiri O, Egan K, Lennon Á, Kevane B, Murphy S, Ní Áinle F, Maguire PB. Proteomic analysis of extracellular vesicle cargoes mirror the cardioprotective effects of rivaroxaban in patients with venous thromboembolism. Proteomics Clin Appl 2024; 18:e202300014. [PMID: 38193270 DOI: 10.1002/prca.202300014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 12/01/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Venous thromboembolism (VTE) remains a significant cause of morbidity and mortality worldwide. Rivaroxaban, a direct oral factor Xa inhibitor, mediates anti-inflammatory and cardiovascular-protective effects besides its well-established anticoagulant properties; yet, these remain poorly characterized. Extracellular vesicles (EVs) are considered proinflammatory messengers regulating a myriad of (patho)physiological processes and may be highly relevant to the pathophysiology of VTE. The effects of Rivaroxaban on circulating EVs in VTE patients remain unknown. We have established that differential EV biosignatures are found in patients with non-valvular atrial fibrillation anticoagulated with Rivaroxaban versus warfarin. Here, we investigated whether differential proteomic profiles of circulating EVs could also be found in patients with VTE. METHODS AND RESULTS We performed comparative label-free quantitative proteomic profiling of enriched plasma EVs from VTE patients anticoagulated with either Rivaroxaban or warfarin using a tandem mass spectrometry approach. Of the 182 quantified proteins, six were found to be either exclusive to, or enriched in, Rivaroxaban-treated patients. Intriguingly, these proteins are involved in negative feedback regulation of inflammatory and coagulation pathways, suggesting that EV proteomic signatures may reflect both Rivaroxaban's anti-coagulatory and anti-inflammatory potential. CONCLUSIONS These differences suggest Rivaroxaban may have pleiotropic effects, supporting the reports of its emerging anti-inflammatory and cardiovascular-protective characteristics relative to warfarin.
Collapse
Affiliation(s)
- Luisa Weiss
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Wido Uhrig
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Sarah Kelliher
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Paulina B Szklanna
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Tadhg Prendiville
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Shane P Comer
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Osasere Edebiri
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Karl Egan
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Áine Lennon
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Barry Kevane
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sean Murphy
- Department of Stroke Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnuala Ní Áinle
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Patricia B Maguire
- UCD Conway SPHERE Research group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Amabebe E, Kumar A, Tatiparthy M, Kammala AK, Taylor BD, Menon R. Cargo exchange between human and bacterial extracellular vesicles in gestational tissues: a new paradigm in communication and immune development. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:297-328. [PMID: 39698538 PMCID: PMC11648491 DOI: 10.20517/evcna.2024.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/25/2024] [Accepted: 06/05/2024] [Indexed: 12/20/2024]
Abstract
Host-bacteria and bacteria-bacteria interactions can be facilitated by extracellular vesicles (EVs) secreted by both human and bacterial cells. Human and bacterial EVs (BEVs) propagate and transfer immunogenic cargos that may elicit immune responses in nearby or distant recipient cells/tissues. Hence, direct colonization of tissues by bacterial cells is not required for immunogenic stimulation. This phenomenon is important in the feto-maternal interface, where optimum tolerance between the mother and fetus is required for a successful pregnancy. Though the intrauterine cavity is widely considered sterile, BEVs from diverse sources have been identified in the placenta and amniotic cavity. These BEVs can be internalized by human cells, which may help them evade host immune surveillance. Though it appears logical, whether bacterial cells internalize human EVs or human EV cargo is yet to be determined. However, the presence of BEVs in placental tissues or amniotic cavity is believed to trigger a low-grade immune response that primes the fetal immune system for ex-utero survival, but is insufficient to disrupt the progression of pregnancy or cause immune intolerance required for adverse pregnancy events. Nevertheless, the exchange of bioactive cargos between human and BEVs, and the mechanical underpinnings and health implications of such interactions, especially during pregnancy, are still understudied. Therefore, while focusing on the feto-maternal interface, we discussed how human cells take up BEVs and whether bacterial cells take up human EVs or their cargo, the exchange of cargos between human and BEVs, host cell (feto-maternal) inflammatory responses to BEV immunogenic stimulation, and associations of these interactions with fetal immune priming and adverse reproductive outcomes such as preeclampsia and preterm birth.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramkumar Menon
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| |
Collapse
|
15
|
Cheung SWY, Chamley LW, Barrett CJ, Lau SYS. Extracellular vesicles and their effect on vascular haemodynamics: a systematic review. Hypertens Res 2024; 47:1588-1606. [PMID: 38600279 PMCID: PMC11150158 DOI: 10.1038/s41440-024-01659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/03/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Extracellular vesicles (EVs) are released from all cell types studied to date and act as intercellular communicators containing proteins, nucleic acids and lipid cargos. They have been shown to be involved in maintaining homoeostasis as well as playing a role in the development of pathology including hypertension and cardiovascular disease. It is estimated that there is 109-1010 circulating EVs/mL in the plasma of healthy individuals derived from various sources. While the effect of EVs on vascular haemodynamic parameters will be dependent on the details of the model studied, we systematically searched and summarized current literature to find patterns in how exogenously injected EVs affected vascular haemodynamics. Under homoeostatic conditions, evidence from wire and pressure myography data demonstrate that injecting isolated EVs derived from cell types found in blood and blood vessels resulted in the impairment of vasodilation in blood vessels ex vivo. Impaired vasodilation was also observed in rodents receiving intravenous injections of human plasma EVs from cardiovascular diseases including valvular heart disease, acute coronary syndrome, myocardial infarction and end stage renal disease. When EVs were derived from models of metabolic syndromes, such as diabetes, these EVs enhanced vasoconstriction responses in blood vessels ex vivo. There were fewer publications that assessed the effect of EVs in anaesthetised or conscious animals to confirm whether effects on the vasculature observed in ex vivo studies translated into alterations in vascular haemodynamics in vivo. In the available conscious animal studies, the in vivo data did not always align with the ex vivo data. This highlights the importance of in vivo work to determine the effects of EVs on the integrative vascular haemodynamics.
Collapse
Affiliation(s)
- Sharon W Y Cheung
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Hub for Extracellular Vesicle Investigations, The University of Auckland, Auckland, New Zealand
| | - Carolyn J Barrett
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Sien Yee S Lau
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
16
|
Afzal A, Khan M, Gul Z, Asif R, Shahzaman S, Parveen A, Imran M, Khawar MB. Extracellular Vesicles: the Next Frontier in Pregnancy Research. Reprod Sci 2024; 31:1204-1214. [PMID: 38151656 DOI: 10.1007/s43032-023-01434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
Extracellular vehicles (EVs) have been involved in several aspects of pregnancy, including endometrial receptivity, embryo implantation, and embryo-maternal communication showing them associated with pregnancy disorders, such as preeclampsia, gestational diabetes mellitus, and preterm birth. Further research is warranted to fully comprehend the exact pathophysiological roles of EVs and to develop new therapies targeting EVs thereby improving pregnancy outcomes. Herein, we review the recent knowledge on the multifaceted roles of EVs during pregnancy and address the majority of the molecular interactions between EVs, maternal, and fetal cells with an emphasis on disorders of pregnancy under the influence of EVs. Moreover, we also discuss its applications in clinical trials followed by prospects.
Collapse
Affiliation(s)
- Ali Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Madeeha Khan
- College of Allied Health Sciences, Akhtar Saeed Medical and Dental College, Lahore, Pakistan
| | - Zaman Gul
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Rameen Asif
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Asia Parveen
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Muhammad Imran
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Babar Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Applied Molecular Biology & Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan.
| |
Collapse
|
17
|
Graf I, Urbschat C, Arck PC. The 'communicatome' of pregnancy: spotlight on cellular and extravesicular chimerism. EMBO Mol Med 2024; 16:700-714. [PMID: 38467841 PMCID: PMC11018796 DOI: 10.1038/s44321-024-00045-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/13/2024] Open
Abstract
Communication via biological mediators between mother and fetus are key to reproductive success and offspring's future health. The repertoire of mediators coding signals between mother and fetus is broad and includes soluble factors, membrane-bound particles and immune as well as non-immune cells. Based on the emergence of technological advancements over the last years, considerable progress has been made toward deciphering the "communicatome" between fetus and mother during pregnancy and even after birth. In this context, pregnancy-associated chimerism has sparked the attention among immunologists, since chimeric cells-although low in number-are maintained in the allogeneic host (mother or fetus) for years after birth. Other non-cellular structures of chimerism, e.g. extracellular vesicles (EVs), are increasingly recognized as modulators of pregnancy outcome and offspring's health. We here discuss the origin, distribution and function of pregnancy-acquired microchimerism and chimeric EVs in mother and offspring. We also highlight the pioneering concept of maternal microchimeric cell-derived EVs in offspring. Such insights expand the understanding of pregnancy-associated health or disease risks in mother and offspring.
Collapse
Affiliation(s)
- Isabel Graf
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christopher Urbschat
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Shan Y, Hou B, Wang J, Chen A, Liu S. Exploring the role of exosomal MicroRNAs as potential biomarkers in preeclampsia. Front Immunol 2024; 15:1385950. [PMID: 38566996 PMCID: PMC10985148 DOI: 10.3389/fimmu.2024.1385950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
The complex pathogenesis of preeclampsia (PE), a significant contributor to maternal and neonatal mortality globally, is poorly understood despite substantial research. This review explores the involvement of exosomal microRNAs (exomiRs) in PE, focusing on their impact on the protein kinase B (AKT)/hypoxia-inducible factor 1-α (HIF1α)/vascular endothelial growth factor (VEGF) signaling pathway as well as endothelial cell proliferation and migration. Specifically, this article amalgamates existing evidence to reveal the pivotal role of exomiRs in regulating mesenchymal stem cell and trophoblast function, placental angiogenesis, the renin-angiotensin system, and nitric oxide production, which may contribute to PE etiology. This review emphasizes the limited knowledge regarding the role of exomiRs in PE while underscoring the potential of exomiRs as non-invasive biomarkers for PE diagnosis, prediction, and treatment. Further, it provides valuable insights into the mechanisms of PE, highlighting exomiRs as key players with clinical implications, warranting further exploration to enhance the current understanding and the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bo Hou
- Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingli Wang
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aiping Chen
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
20
|
Farrelly R, Kennedy MG, Spencer R, Forbes K. Extracellular vesicles as markers and mediators of pregnancy complications: gestational diabetes, pre-eclampsia, preterm birth and fetal growth restriction. J Physiol 2023; 601:4973-4988. [PMID: 37070801 PMCID: PMC11497252 DOI: 10.1113/jp282849] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/13/2023] [Indexed: 04/19/2023] Open
Abstract
In high income countries, approximately 10% of pregnancies are complicated by pre-eclampsia (PE), preterm birth (PTB), fetal growth restriction (FGR) and/or macrosomia resulting from gestational diabetes (GDM). Despite the burden of disease this places on pregnant people and their newborns, there are still few, if any, effective ways of preventing or treating these conditions. There are also gaps in our understanding of the underlying pathophysiologies and our ability to predict which mothers will be affected. The placenta plays a crucial role in pregnancy, and alterations in placental structure and function have been implicated in all of these conditions. As extracellular vesicles (EVs) have emerged as important molecules in cell-to-cell communication in health and disease, recent research involving maternal- and placental-derived EV has demonstrated their potential as predictive and diagnostic biomarkers of obstetric disorders. This review will consider how placental and maternal EVs have been investigated in pregnancies complicated by PE, PTB, FGR and GDM and aims to highlight areas where further research is required to enhance the management and eventual treatment of these pathologies.
Collapse
Affiliation(s)
- Rachel Farrelly
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | | | - Rebecca Spencer
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Karen Forbes
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
21
|
Rao A, Shinde U, Das DK, Balasinor N, Madan T. Early prediction of pre-eclampsia using circulating placental exosomes: Newer insights. Indian J Med Res 2023; 158:385-396. [PMID: 37987999 DOI: 10.4103/ijmr.ijmr_2143_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 11/22/2023] Open
Abstract
Pre-eclampsia (PE), a multifactorial de novo hypertensive pregnancy disorder, is one of the leading causes of foeto-maternal morbidity and mortality. Currently, antihypertensive drugs are the first-line therapy for PE and evidence suggests that low-dose aspirin initiated early in high risk pregnancies may reduce the risk of development or severity of PE. However, an early prediction of this disorder remains an unmet clinical challenge. Several potential serum biomarkers associated with maternal immunoregulation and placental angiogenesis have been evaluated but are ineffective and inconsistent for early prediction. Although placental biomarkers would be more specific and sensitive in predicting the risk of PE, accessing the placenta during pregnancy is not feasible. Circulating placental exosomes (pEXO), originating from foeto-maternal interface, are being evaluated as the placenta's surrogate and the best source of non-invasive placental biomarkers. pEXO appear in the maternal circulation starting from six weeks of gestation and its dynamic biological cargo across pregnancy is associated with successful pregnancy outcomes. Therefore, monitoring changes in pEXO expression profiles could provide new insights into the prediction, diagnosis and treatment of PE. This narrative review comprehensively summarizes the available literature on the candidate predictive circulating biomarkers evaluated for PE to date. In particular, the review elucidates the current knowledge of distinct molecular signatures emanating from pEXO in pre-eclamptic women to support the discovery of novel early predictive biomarkers for effective intervention and management of the disease.
Collapse
Affiliation(s)
- Aishwarya Rao
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Uma Shinde
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Dhanjit Kumar Das
- Department of Stem Cell Biology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Nafisa Balasinor
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Taruna Madan
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
22
|
Paul N, Sultana Z, Fisher JJ, Maiti K, Smith R. Extracellular vesicles- crucial players in human pregnancy. Placenta 2023; 140:30-38. [PMID: 37531747 DOI: 10.1016/j.placenta.2023.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 08/04/2023]
Abstract
Extracellular vesicles (EVs) are lipid-bilayer enclosed membrane vesicles released by cells in physiological and pathological states. EVs are generated and released through a variety of pathways and mediate cellular communication by carrying and transferring signals to recipient cells. EVs are specifically loaded with proteins, nucleic acids (RNAs and DNA), enzymes and lipids, and carry a range of surface proteins and adhesion molecules. EVs contribute to intercellular signalling, development, metabolism, tissue homeostasis, antigen presentation, gene expression and immune regulation. EVs have been categorised into three different subgroups based on their size: exosomes (30-150 nm), microvesicles (100-1000 nm) and apoptotic bodies (1-5 μm). The status of the cells of origin of EVs influences their biology, heterogeneity and functions. EVs, especially exosomes, have been studied for their potential roles in feto-maternal communication and impacts on normal pregnancy and pregnancy disorders. This review presents an overview of EVs, emphasising exosomes and microvesicles in a general context, and then focusing on the roles of EVs in human pregnancy and their potential as diagnostics for adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Nilanjana Paul
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia.
| | - Zakia Sultana
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia.
| | - Joshua J Fisher
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia.
| | | | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia.
| |
Collapse
|
23
|
Kuebler WM, William N, Post M, Acker JP, McVey MJ. Extracellular vesicles: effectors of transfusion-related acute lung injury. Am J Physiol Lung Cell Mol Physiol 2023; 325:L327-L341. [PMID: 37310760 DOI: 10.1152/ajplung.00040.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Respiratory transfusion reactions represent some of the most severe adverse reactions related to receiving blood products. Of those, transfusion-related acute lung injury (TRALI) is associated with elevated morbidity and mortality. TRALI is characterized by severe lung injury associated with inflammation, pulmonary neutrophil infiltration, lung barrier leak, and increased interstitial and airspace edema that cause respiratory failure. Presently, there are few means of detecting TRALI beyond clinical definitions based on physical examination and vital signs or preventing/treating TRALI beyond supportive care with oxygen and positive pressure ventilation. Mechanistically, TRALI is thought to be mediated by the culmination of two successive proinflammatory hits, which typically comprise a recipient factor (1st hit-e.g., systemic inflammatory conditions) and a donor factor (2nd hit-e.g., blood products containing pathogenic antibodies or bioactive lipids). An emerging concept in TRALI research is the contribution of extracellular vesicles (EVs) in mediating the first and/or second hit in TRALI. EVs are small, subcellular, membrane-bound vesicles that circulate in donor and recipient blood. Injurious EVs may be released by immune or vascular cells during inflammation, by infectious bacteria, or in blood products during storage, and can target the lung upon systemic dissemination. This review assesses emerging concepts such as how EVs: 1) mediate TRALI, 2) represent targets for therapeutic intervention to prevent or treat TRALI, and 3) serve as biochemical biomarkers facilitating TRALI diagnosis and detection in at-risk patients.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin, Berlin, Germany
- Keenan Research Centre, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Nishaka William
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Martin Post
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Jason P Acker
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Edmonton, Alberta, Canada
| | - Mark J McVey
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Chen Q, He J, Liu H, Huang Q, Wang S, Yin A, Chen S, Shen X, Xiao Y, Hu H, Jiang J, Chen W, Wang S, Huang Z, Li J, Peng Y, Wang X, Yang X, Wang Z, Zhong M. Small extracellular vesicles-transported lncRNA TDRKH-AS1 derived from AOPPs-treated trophoblasts initiates endothelial cells pyroptosis through PDIA4/DDIT4 axis in preeclampsia. J Transl Med 2023; 21:496. [PMID: 37488572 PMCID: PMC10364420 DOI: 10.1186/s12967-023-04346-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Substantial studies have demonstrated that oxidative stress placenta and endothelial injury are considered to inextricably critical events in the pathogenesis of preeclampsia (PE). Systemic inflammatory response and endothelial dysfunction are induced by the circulating factors released from oxidative stress placentae. As a novel biomarker of oxidative stress, advanced oxidation protein products (AOPPs) levels are strongly correlated with PE characteristics. Nevertheless, the molecular mechanism underlying the effect of factors is still largely unknown. METHODS With the exponential knowledge on the importance of placenta-derived extracellular vesicles (pEVs), we carried out lncRNA transcriptome profiling on small EVs (sEVs) secreted from AOPPs-treated trophoblast cells and identified upregulated lncRNA TDRKH-AS1 as a potentially causative factor for PE. We isolated and characterized sEVs from plasma and trophoblast cells by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting. The expression and correlation of lncRNA TDRKH-AS1 were evaluated using qRT-PCR in plasmatic sEVs and placentae from patients. Pregnant mice injected with TDRKH-AS1-riched trophoblast sEVs was performed to detect the TDRKH-AS1 function in vivo. To investigate the potential effect of sEVs-derived TDRKH-AS1 on endothelial function in vitro, transcriptome sequencing, scanning electron Microscopy (SEM), immunofluorescence, ELISA and western blotting were conducted in HUVECs. RNA pulldown, mass spectrometry, RNA immunoprecipitation (RIP), chromatin isolation by RNA purification (ChIRP) and coimmunoprecipitation (Co-IP) were used to reveal the latent mechanism of TDRKH-AS1 on endothelial injury. RESULTS The expression level of TDRKH-AS1 was significantly increased in plasmatic sEVs and placentae from patients, and elevated TDRKH-AS1 in plasmatic sEVs was positively correlated with clinical severity of the patients. Moreover, pregnant mice injected with TDRKH-AS1-riched trophoblast sEVs exhibited a hallmark feature of PE with increased blood pressure and systemic inflammatory responses. Pyroptosis, an inflammatory form of programmed cell death, is involved in the development of PE. Indeed, our in vitro study indicated that sEVs-derived TDRKH-AS1 secreted from AOPPs-induced trophoblast elevated DDIT4 expression levels to trigger inflammatory response of pyroptosis in endothelial cells through interacting with PDIA4. CONCLUSIONS Herein, results in the present study supported that TDRKH-AS1 in sEVs isolated from oxidative stress trophoblast may be implicated in the pathogenesis of PE via inducing pyroptosis and aggravating endothelial dysfunction.
Collapse
Affiliation(s)
- Qian Chen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiexing He
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haihua Liu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyu Huang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuoshi Wang
- Department of Obstetrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Ailan Yin
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuying Chen
- Department of Obstetrics, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen, 518035, China
| | - Xinyang Shen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanxuan Xiao
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haoyue Hu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayi Jiang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenqian Chen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Song Wang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenqin Huang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Li
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - You Peng
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaocong Wang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinping Yang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China.
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Zhijian Wang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Mei Zhong
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
25
|
Santoyo JM, Noguera JA, Avilés F, Hernández-Caselles T, de Paco-Matallana C, Delgado JL, Cuevas S, Llinás MT, Hernández I. Pravastatin reduces plasma levels of extracellular vesicles in pregnancies at high risk of term preeclampsia. Front Pharmacol 2023; 14:1166123. [PMID: 37426825 PMCID: PMC10323224 DOI: 10.3389/fphar.2023.1166123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: Elevated plasma levels of extracellular vesicles have been associated with impaired placentation, angiogenesis imbalance, intravascular inflammation, and endothelial dysfunction in women with preeclampsia, thus suggesting that circulating vesicles may be a good therapeutic target for the treatment of the disease. Recently, statins have been considered a potential treatment for the prevention of preeclampsia because of their pleiotropic effects, including the improvement of endothelial dysfunction and inhibition of inflammatory responses. However, the effects of these drugs on circulating vesicles concentration in women at risk of preeclampsia have not been established. Herein, we aimed to assess the effects of pravastatin on circulating extracellular vesicle generation in women at high risk of term preeclampsia. Methods: In a sample of 68 singleton pregnant women participating in the multicenter, double-blind, placebo-controlled STATIN trial (Nº EducraCT 2016-005206-19 ISRCTN), 35 women received a placebo and 33 women received a 20 mg/day dose of pravastatin for approximately 3 weeks (from 35 to 37 weeks of gestation until delivery). Large extracellular vesicles were characterized and quantified by flow cytometry using annexin V and cell-specific antibodies directed against platelet, endothelial, leukocyte, and syncytiotrophoblast cell surface markers. Results: In women who received the placebo, a significant increase in the plasma levels of large extracellular vesicles from platelets (34%, p < 0.01), leukocytes (33%, p < 0.01), monocytes (60%, p < 0.01), endothelial cells (40%, p < 0.05), and syncytiotrophoblast cells (22%, p < 0.05) were observed. However, treatment with pravastatin significantly reduced the plasma levels of large extracellular vesicles from platelets (42%, p < 0.001), leukocytes (25%, p < 0.001), monocytes (61%, p < 0.001), endothelial cells (69%, p < 0.001), activated endothelial cells (55%, p < 0.001), and syncytiotrophoblast cells (44%, p < 0.001). Discussion: These results indicate that pravastatin reduces the levels of activated cell-derived membrane vesicles from the maternal vasculature, blood, and placental syncytiotrophoblast of women at high risk of term preeclampsia, suggesting that this statin may be beneficial in reducing endothelial dysfunction and pro-inflammatory and pro-coagulatory state characteristics of the disease.
Collapse
Affiliation(s)
- Jean Michell Santoyo
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - José Antonio Noguera
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Francisco Avilés
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Trinidad Hernández-Caselles
- Department of Biochemistry and Molecular Biology “B” and Immunology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Catalina de Paco-Matallana
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Juan Luis Delgado
- Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Santiago Cuevas
- Molecular Inflammation Group, Institute of Biomedical Research (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - M. Teresa Llinás
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Isabel Hernández
- Department of Physiology, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| |
Collapse
|
26
|
Beetler DJ, Di Florio DN, Bruno KA, Ikezu T, March KL, Cooper LT, Wolfram J, Fairweather D. Extracellular vesicles as personalized medicine. Mol Aspects Med 2023; 91:101155. [PMID: 36456416 PMCID: PMC10073244 DOI: 10.1016/j.mam.2022.101155] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/14/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles (EVs) are released from all cells in the body, forming an important intercellular communication network that contributes to health and disease. The contents of EVs are cell source-specific, inducing distinct signaling responses in recipient cells. The specificity of EVs and their accumulation in fluid spaces that are accessible for liquid biopsies make them highly attractive as potential biomarkers and therapies for disease. The duality of EVs as favorable (therapeutic) or unfavorable (pathological) messengers is context dependent and remains to be fully determined in homeostasis and various disease states. This review describes the use of EVs as biomarkers, drug delivery vehicles, and regenerative therapeutics, highlighting examples involving viral infections, cancer, and neurological diseases. There is growing interest to provide personalized therapy based on individual patient and disease characteristics. Increasing evidence suggests that EV biomarkers and therapeutic approaches are ideal for personalized medicine due to the diversity and multifunctionality of EVs.
Collapse
Affiliation(s)
- Danielle J Beetler
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Damian N Di Florio
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA; Center for Regenerative Medicine, University of Florida, Gainesville, FL, 32611, USA; Division of Cardiology, University of Florida, Gainesville, FL, 32611, USA
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Keith L March
- Center for Regenerative Medicine, University of Florida, Gainesville, FL, 32611, USA; Division of Cardiology, University of Florida, Gainesville, FL, 32611, USA
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - DeLisa Fairweather
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
27
|
Sha M, Zhang S, Beejadhursing R, Sun Y, Qin Y, Chen S, Li W. Extracellular vesicles derived from hypoxic HTR-8/SVneo trophoblast inhibit endothelial cell functions through the miR-150-3p /CHPF pathway. Placenta 2023; 138:21-32. [PMID: 37156185 DOI: 10.1016/j.placenta.2023.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/21/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Endothelial dysfunction is one of the basic pathological changes in pre-eclampsia. Extracellular vesicles (EVs) can transport miRNAs expressed by placental trophoblast cells into endothelial cells. The aim of this study was to explore the differential effects of EVs induced by hypoxic trophoblasts (1%HTR-8-EV) and those derived from normoxic trophoblasts (20%HTR-8-EV) on the regulation of endothelial cell functions. METHODS Normoxia and hypoxia were preconditioned to induce trophoblast cells-derived EVs. The effect of EVs, miRNA, target gene, and their interactions on endothelial cell proliferation, migration, and angiogenesis were determined. Quantitative analysis of miR-150-3p and CHPF were verified by qRT-PCR and western blotting. The binding relationship among EVs pathway was demonstrated by luciferase reporter assay. RESULTS Compared with 20%HTR-8-EV, 1%HTR-8-EV had a suppressive effect on proliferation, migration, and angiogenesis of endothelial cells. The results of miRNA sequencing showed the vital role of miR-150-3p in trophoblast-to-endothelium communication. 1%HTR-8-EV carrying miR-150-3p could move into endothelial cells and target chondroitin polymerizing factor (CHPF) gene. MiR-150-3p inhibited endothelial cell functions by regulating CHPF. In patient-derived placental vascular tissues, there was a similar negative correlating between miR-150-3p and CHPF. DISCUSSION Our findings indicate that extracellular vesicles miR-150-3p derived from hypoxic trophoblasts inhibits endothelial cells proliferation, migration, and angiogenesis by modulating CHPF, illuminating a novel mechanism of hypoxic trophoblasts regulation of endothelial cells and their potential role in PE pathogenesis.
Collapse
Affiliation(s)
- Menghan Sha
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Shunran Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rajluxmee Beejadhursing
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanan Sun
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Qin
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Suhua Chen
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Wei Li
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
28
|
Wu S, Li Q, Liu X, Huang H, Wang G, Zhang C, Meng Y, Yang W. Placental exosomal miR-125b triggered endothelial barrier injury in preeclampsia. Placenta 2023; 137:31-37. [PMID: 37054628 DOI: 10.1016/j.placenta.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/12/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
INTRODUCTION Preeclampsia (PE) is an elusive life-threatening complication of pregnancy, and maternal endothelial dysfunction induced by components from the impaired placenta is a key hallmark of PE. Placenta-derived exosomes in maternal circulation have been correlated with risk of PE, however, the role of exosomes in PE remains to be determined. We hypothesized that placenta-released exosomes link the placental abnormalities with maternal endothelial dysfunction in PE. METHODS Circulating exosomes were collected from plasma samples of preeclamptic patients and normal pregnancies. Endothelial barrier function was examined by transendothelial electrical resistance (TEER) and cell permeability to FITC-dextran assays in human umbilical vein endothelial cells (HUVECs). miR-125b and VE-cadherin gene expression in exosomes and endothelial cells were assessed by qPCR and Western, and the possible post-transcriptional regulation of miR-125b on VE-cadherin was detected by luciferase assay. RESULTS We isolated placenta-derived exosomes in the maternal circulation and found that placenta-derived exosomes from preeclamptic patients (PE-exo) leads to endothelial barrier dysfunction. We then identified decreased expression of VE-cadherin in endothelial cells contribute to the breakdown of the endothelial barrier. Further investigations revealed increased exosomal miR-125b in PE-exo directly inhibited VE-cadherin in HUVECs, thereby mediating the adverse effect of PE-exo on endothelial barrier function. DISCUSSION Placental exosomes link impaired placentation and endothelial dysfunction, thus providing new insight into the pathophysiology of preeclampsia. Exosomal miRNAs derived from placenta contribute to the endothelial dysfunction in PE and could be a promising therapeutic target for PE.
Collapse
Affiliation(s)
- Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| | - Qinghua Li
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; School of Public Health, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Xinlu Liu
- School of Biosciences, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Haiqin Huang
- School of Biosciences, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Guohui Wang
- School of Biosciences, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Cuijuan Zhang
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, Weifang, 261031, Shandong, China
| | - Yuhan Meng
- Center for Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, 261042, Shandong, China
| | - Weiwei Yang
- School of Biosciences, Weifang Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
29
|
Xie L, Ding N, Sheng S, Zhang H, Yin H, Gao L, Zhang H, Ma S, Yang A, Li G, Jiao Y, Shi Q, Jiang Y, Zhang H. Cooperation between NSPc1 and DNA methylation represses HOXA11 expression and promotes apoptosis of trophoblast cells during preeclampsia. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1-13. [PMID: 36815373 PMCID: PMC10157525 DOI: 10.3724/abbs.2023012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/03/2022] [Indexed: 02/05/2023] Open
Abstract
Accumulating evidence has shown that the apoptosis of trophoblast cells plays an important role in the pathogenesis of preeclampsia, and an intricate interplay between DNA methylation and polycomb group (PcG) protein-mediated gene silencing has been highlighted recently. Here, we provide evidence that the expression of nervous system polycomb 1 (NSPc1), a BMI1 homologous polycomb protein, is significantly elevated in trophoblast cells during preeclampsia, which accelerates trophoblast cell apoptosis. Since NSPc1 acts predominantly as a transcriptional inactivator that specifically represses HOXA11 expression in trophoblast cells during preeclampsia, we further show that NSPc1 is required for DNMT3a recruitment and maintenance of the DNA methylation in the HOXA11 promoter in trophoblast cells during preeclampsia. In addition, we find that the interplay of DNMT3a and NSPc1 represses the expression of HOXA11 and promotes trophoblast cell apoptosis. Taken together, these results indicate that the cooperation between NSPc1 and DNMT3a reduces HOXA11 expression in preeclampsia pathophysiology, which provides novel therapeutic approaches for targeted inhibition of trophoblast cell apoptosis during preeclampsia pathogenesis.
Collapse
Affiliation(s)
- Lin Xie
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Ning Ding
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Siqi Sheng
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Honghong Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - He Yin
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- Department of Clinical MedicineNingxia Medical UniversityYinchuan750004China
| | - Lina Gao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- Department of Clinical MedicineNingxia Medical UniversityYinchuan750004China
| | - Hui Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Shengchao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Anning Yang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Yun Jiao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- Department of Infectious DiseasesGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Qing Shi
- Department of GynecologyGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Huiping Zhang
- Department of Medical GeneticsMaternal and Child Health of Hunan ProvinceChangsha410008China
- Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004China
- General Hospital of Ningxia Medical UniversityYinchuan750004China
| |
Collapse
|
30
|
Escudero C, Kupka E, Ibañez B, Sandoval H, Troncoso F, Wikström AK, López-Espíndola D, Acurio J, Torres-Vergara P, Bergman L. Brain Vascular Dysfunction in Mothers and Their Children Exposed to Preeclampsia. Hypertension 2023; 80:242-256. [PMID: 35920147 DOI: 10.1161/hypertensionaha.122.19408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Preeclampsia is a maternal syndrome characterized by the new onset of hypertension and proteinuria after 20 weeks of gestation associated with multisystemic complications, including brain alterations. Indeed, brain complications associated with preeclampsia are the leading direct causes of fetal and maternal morbidity and mortality, especially in low- and middle-income countries. In addition to the well-recognized long-term adverse cardiovascular effects of preeclampsia, women who have had preeclampsia have higher risk of stroke, dementia, intracerebral white matter lesions, epilepsy, and perhaps also cognitive decline postpartum. Furthermore, increasing evidence has also associated preeclampsia with similar cognitive and cerebral disorders in the offspring. However, the mechanistic links between these associations remain unresolved. This article summarizes the current knowledge about the cerebrovascular complications elicited by preeclampsia and the potential pathophysiological mechanisms involved, emphasizing the impaired brain vascular function in the mother and their offspring.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Ellen Kupka
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Sweden (E.K.)
| | - Belen Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.).,Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Chile (P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.).,Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.).,Department of clinical sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
| |
Collapse
|
31
|
Adamova P, Lotto RR, Powell AK, Dykes IM. Are there foetal extracellular vesicles in maternal blood? Prospects for diagnostic biomarker discovery. J Mol Med (Berl) 2023; 101:65-81. [PMID: 36538060 PMCID: PMC9977902 DOI: 10.1007/s00109-022-02278-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 03/02/2023]
Abstract
Prenatal diagnosis of congenital disease improves clinical outcomes; however, as many as 50% of congenital heart disease cases are missed by current ultrasound screening methods. This indicates a need for improved screening technology. Extracellular vesicles (EVs) have attracted enormous interest in recent years for their potential in diagnostics. EVs mediate endocrine signalling in health and disease and are known to regulate aspects of embryonic development. Here, we critically evaluate recent evidence suggesting that EVs released from the foetus are able to cross the placenta and enter the maternal circulation. Furthermore, EVs from the mother appear to be transported in the reverse direction, whilst the placenta itself acts as a source of EVs. Experimental work utilising rodent models employing either transgenically encoded reporters or application of fluorescent tracking dyes provide convincing evidence of foetal-maternal crosstalk. This is supported by clinical data demonstrating expression of placental-origin EVs in maternal blood, as well as limited evidence for the presence of foetal-origin EVs. Together, this work raises the possibility that foetal EVs present in maternal blood could be used for the diagnosis of congenital disease. We discuss the challenges faced by researchers in translating these basic science findings into a clinical non-invasive prenatal test.
Collapse
Affiliation(s)
- Petra Adamova
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Robyn R Lotto
- Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.,School of Nursing and Allied Health, Liverpool John Moores University, Tithebarn St, Liverpool, L2 2ER, UK
| | - Andrew K Powell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Iain M Dykes
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK. .,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
32
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Wang Y, Bai X, Guo X, Gao X, Chen Y, Li H, Fan W, Han C. Bioinformatics analysis combined with clinical sample screening reveals that leptin may be a biomarker of preeclampsia. Front Physiol 2023; 13:1031950. [PMID: 36685185 PMCID: PMC9846503 DOI: 10.3389/fphys.2022.1031950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Preeclampsia (PE) is a gestational hypertensive disease with unclear pathogenesis. This study aimed to identify the genes that play an important role in determining the pathogenesis of PE using bioinformatics analysis and fundamental researches. Materials and methods: Datasets from the Gene Expression Omnibus (GEO) database were used to screen for differentially expressed genes (DEGs). The NCBI, SangerBox, and other databases were used to analyze the functions of the DEGs. Targetscan7, miRWalk, ENCORI, DIANA TOOLS, CircBank databases, and the Cytoscape tool were used to construct the lncRNA/circRNA-miRNA- LEP network. SRAMP, RPISeq, RBPsuite, and catRPAID were used to analyze the RNA modifications of LEP. Immune cell infiltration was analyzed using the dataset GSE75010. Placental tissues from normal pregnant women and PE patients were collected, screened for gene expression using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. The results were further verified in HTR-8/SVneo cell line hypoxia model and PE mouse model. Results: Our analyses revealed that LEP was significantly upregulated in eight datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses indicated that LEP was involved in the JAK/STAT signaling pathway, angiogenesis, and placental development. Immune cell infiltration analysis showed that M1 and M2 macrophages differed between normal pregnancies and those in PE patients. A competing endogenous RNA (ceRNA) network was constructed, and proteins interacting with LEP were identified. RNA modification sites of LEP were also identified. Finally, the overexpression of LEP in PE was confirmed in clinical samples, HTR-8/SVneo cell line and PE mouse model. Conclusion: Our results indicate that LEP overexpression is associated with PE and may be a potential diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Yajuan Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuening Bai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoli Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuanyuan Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenjun Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Cha Han,
| |
Collapse
|
34
|
Placental extracellular vesicles in maternal-fetal communication during pregnancy. Biochem Soc Trans 2022; 50:1785-1795. [DOI: 10.1042/bst20220734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
For several years, a growing number of studies have highlighted the pivotal role of placental extracellular vesicles (EVs) throughout pregnancy. These membrane nanovesicles, heterogeneous in nature, composition and origin, are secreted by several trophoblastic cell types and are found in both the maternal and fetal compartments. They can be uptaken by recipient cells and drive a wide variety of physiological and pathological processes. In this review, we provide an overview of the different described roles of placental EVs in various aspects of normal pregnancy, from placenta establishment to maternal immune tolerance towards the fetus and protection against viral infections. In the second part, we present selected examples of pathological pregnancies in which placental EVs are involved, such as gestational diabetes mellitus, pre-eclampsia, and congenital infections. Since the abundance and/or composition of placental EVs is deregulated in maternal serum during pathological pregnancies, this makes them interesting candidates as non-invasive biomarkers for gestational diseases and opens a wide field of translational perspectives.
Collapse
|
35
|
Guo H, Zhang Y, Chu Y, Yang S, Zhang J, Qiao R. Recombinant protein diannexin prevents preeclampsia-like symptoms in a pregnant mouse model via reducing the release of microparticles. Front Med 2022; 16:919-931. [PMID: 36331793 DOI: 10.1007/s11684-021-0918-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/21/2021] [Indexed: 11/06/2022]
Abstract
Preeclampsia (PE) is characterized by placenta-mediated pregnancy complication. The only effective treatment for PE is the delivery of the placenta. However, this treatment may cause preterm birth and neonatal death. Therefore, preventing PE is needed. The mechanism of PE involves abnormal placentation, which leads to the release of anti-angiogenic and inflammatory mediators into maternal circulation. These mediators contribute to systemic vascular dysfunction, inflammatory responses, and excessive thrombin generation. Microparticles (MPs) are reportedly involved in PE by promoting the thromboinflammatory response. This study describes a strategy to prevent PE by reducing MP release using the recombinant protein, diannexin. Results showed that the patients with PE had elevated MP number and procoagulant activity and increased NLRP3 inflammasome activation. Additionally, diannexin remarkably reduced the release of MPs from activated cells by binding to phosphatidylserine exposed on the surface of activated cells. Moreover, in vivo results showed that diannexin could prevent PE-like symptoms by decreasing MPs and NLRP3 inflammasome activation in pregnant mice. Furthermore, diannexin effectively inhibited trophoblast cell activation and NLRP3 inflammasome activation in vitro. These findings suggested that diannexin inhibited MP release and might be an effective therapeutic strategy for preventing PE.
Collapse
Affiliation(s)
- Han Guo
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yuncong Zhang
- Department of Clinical Laboratory, Peking University International Hospital, Beijing, 102206, China
| | - Yaxin Chu
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuo Yang
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Jie Zhang
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Rui Qiao
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
36
|
Ortega MA, Fraile-Martínez O, García-Montero C, Paradela A, Asunción Sánchez-Gil M, Rodriguez-Martin S, De León-Luis JA, Pereda-Cerquella C, Bujan J, Guijarro LG, Alvarez-Mon M, García-Honduvilla N. Unfolding the role of placental-derived Extracellular Vesicles in Pregnancy: From homeostasis to pathophysiology. Front Cell Dev Biol 2022; 10:1060850. [PMID: 36478738 PMCID: PMC9720121 DOI: 10.3389/fcell.2022.1060850] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/02/2022] [Indexed: 08/11/2023] Open
Abstract
The human placenta is a critical structure with multiple roles in pregnancy, including fetal nutrition and support, immunological, mechanical and chemical barrier as well as an endocrine activity. Besides, a growing body of evidence highlight the relevance of this organ on the maternofetal wellbeing not only during gestation, but also from birth onwards. Extracellular vesicles (EVs) are complex macromolecular structures of different size and content, acting as carriers of a diverse set of molecules and information from donor to recipient cells. Since its early development, the production and function of placental-derived EVs are essential to ensure an adequate progress of pregnancy. In turn, the fetus receives and produce their own EVs, highlighting the importance of these components in the maternofetal communication. Moreover, several studies have shown the clinical relevance of EVs in different obstetric pathologies such as preeclampsia, infectious diseases or gestational diabetes, among others, suggesting that they could be used as pathophysiological biomarkers of these diseases. Overall, the aim of this article is to present an updated review of the published basic and translational knowledge focusing on the role of placental-derived EVs in normal and pathological pregnancies. We suggest as well future lines of research to take in this novel and promising field.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | | | - María Asunción Sánchez-Gil
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| | - Sonia Rodriguez-Martin
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Service of Pediatric, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Juan A. De León-Luis
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Ma-drid, Madrid, Spain
| | - Claude Pereda-Cerquella
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Luis G. Guijarro
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), Department of System Biology, University of Alcalá, Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), University Hospital Príncipe de Asturias, Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| |
Collapse
|
37
|
Gao Y, Li K, Li X, Li Q, Wang J, Zhang S, Zhang J. Exploration of cerebral vasospasm from the perspective of microparticles. Front Neurosci 2022; 16:1013437. [PMID: 36389239 PMCID: PMC9650082 DOI: 10.3389/fnins.2022.1013437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/14/2022] [Indexed: 02/28/2024] Open
Abstract
Cerebral vasospasm is a frequently encountered clinical problem, especially in patients with traumatic brain injury and subarachnoid hemorrhage. Continued cerebral vasospasm can cause cerebral ischemia, even infarction and delayed ischemic neurologic deficits. It significantly affects the course of the disease and the outcome of the patient. However, the underlying mechanism of cerebral vasospasm is still unclear. Recently, increasing studies focus on the pathogenic mechanism of microparticles. It has been found that microparticles have a non-negligible role in promoting vasospasm. This research aims to summarize the dynamics of microparticles in vivo and identify a causal role of microparticles in the occurrence and development of cerebral vasospasm. We found that these various microparticles showed dynamic characteristics in body fluids and directly or indirectly affect the cerebral vasospasm or prompt it. Due to the different materials carried by microparticles from different cells, there are also differences in the mechanisms that lead to abnormal vasomotor. We suggest that microparticle scavengers might be a promising therapeutic target against microparticles associated complications.
Collapse
Affiliation(s)
- Yalong Gao
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Kai Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotian Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qifeng Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiwei Wang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Shu Zhang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
38
|
Liu X, Fei H, Yang C, Wang J, Zhu X, Yang A, Shi Z, Jin X, Yang F, Wu D, Jiang L, Zhang S. Trophoblast-Derived Extracellular Vesicles Promote Preeclampsia by Regulating Macrophage Polarization. Hypertension 2022; 79:2274-2287. [PMID: 35993233 DOI: 10.1161/hypertensionaha.122.19244] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Systemic inflammation caused by dysfunctional macrophages is a crucial pathogenetic event in preeclampsia (PE). Trophoblast-derived extracellular vesicles (T-EVs) are potent immune cell signaling modulators in pregnancy. Herein, we aimed to investigate T-EVs' effect and mechanism on macrophage polarization and its role in PE pathogenesis, which remain unclear. METHODS Flow cytometry and immunochemistry were used to determine placental macrophage phenotypes. T-EVs were immuno-isolated via placental alkaline phosphatase antibody and identified by transmission electron microscopy and nanoparticle tracking analysis. Quantitative real-time polymerase chain reaction and flow cytometry were used to examine the effects of T-EVs on macrophage polarization, and correlation analysis of T-EVs lipidomics and macrophages transcriptome were performed to explore how T-EVs modulate macrophages. Animal experiments were established to investigate the relationship among PE, T-EVs, and macrophages. RESULTS Macrophages shift from the M2 to M1 phenotype in the preeclamptic placenta. Also, T-EVs from women with PE (PE-EVs) significantly upregulated M1 gene markers and significantly downregulated CD163 expression in macrophages compared with T-EVs in women with normal pregnancies (NP-EVs). Mechanistically, correlation analysis with T-EVs lipidome and the transcriptome of macrophages treated with PE-EVs or NP-EVs indicated that 37 lipids altered in PE-EVs considerably affected classical inflammatory biological pathways in macrophages. Finally, animal experiments revealed that PE-EVs triggered PE-like symptoms in pregnant mice, which were alleviated after macrophage depletion. CONCLUSIONS T-EVs from women with PE could promote preeclampsia by inducing macrophage imbalance polarization, signifying a potential novel interventional target for the prevention and management of PE.
Collapse
Affiliation(s)
- Xiu Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Haiyi Fei
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Cuiyu Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Jianmin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Xiaohong Zhu
- Department of Obstetrics and Gynecology, Affiliated Xiao Shan Hospital, Hangzhou Normal University, China (X.Z.)
| | - Anran Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Zhan Shi
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Xiaoying Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Fei Yang
- Chronic Disease Research Institute, The Children's Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, China (F.Y.)
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China (D.W.)
| | - Lingling Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.).,Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, China (X.L., H.F., C.Y., J.W., A.Y., Z.S., X.J., L.J., S.Z.)
| |
Collapse
|
39
|
Wang Z, Zibrila Abdoulaye I, Liu J, Li C. Trophoblast necroptosis in Preeclampsia: The role of fetus-derived exosomal microRNAs. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
40
|
Listeria monocytogenes Infection Alters the Content and Function of Extracellular Vesicles Produced by Trophoblast Stem Cells. Infect Immun 2022; 90:e0034722. [PMID: 36154271 DOI: 10.1128/iai.00347-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Placental immunity is critical for fetal health during pregnancy, as invading pathogens spread from the parental blood to the fetus through this organ. However, inflammatory responses in the placenta can adversely affect both the fetus and the pregnant person, and the balance between protective placental immune response and detrimental inflammation is poorly understood. Extracellular vesicles (EVs) are membrane-enclosed vesicles that play a critical role in placental immunity. EVs produced by placental trophoblasts mediate immune tolerance to the fetus and to the placenta itself, but these EVs can also activate detrimental inflammatory responses. The regulation of these effects is not well characterized, and the role of trophoblast EVs (tEVs) in the response to infection has yet to be defined. The Gram-positive bacterial pathogen Listeria monocytogenes infects the placenta, serving as a model to study tEV function in this context. We investigated the effect of L. monocytogenes infection on the production and function of tEVs, using a trophoblast stem cell (TSC) model. We found that tEVs from infected TSCs can induce the production of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α) in recipient cells. Surprisingly, this tEV treatment could confer increased susceptibility to subsequent L. monocytogenes infection, which has not been reported previously as an effect of EVs. Proteomic analysis and RNA sequencing revealed that tEVs from infected TSCs had altered cargo compared with those from uninfected TSCs. However, no L. monocytogenes proteins were detected in tEVs from infected TSCs. Together, these results suggest an immunomodulatory role for tEVs during prenatal infection.
Collapse
|
41
|
Correlation of Serum Chemokine (C-C Motif) Ligand 21 and Heat Shock Protein 90 with Preeclampsia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2156424. [PMID: 35966726 PMCID: PMC9365573 DOI: 10.1155/2022/2156424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
Objective The study aimed to explore the correlation of serum chemokine (C-C motif) ligand 21 (CCL21) and heat shock protein 90 (Hsp90) with preeclampsia (PE). Methods Between June 2021 and June 2022, 50 pregnant women with PE were included in the PE group, and 50 healthy pregnant women were included in the control group. The serum levels of CCL21 and Hsp90 were compared between the two groups. Results PE patients showed significantly higher levels of CCL21 and Hsp90 than healthy pregnant women (P < 0.05). Correlation analysis showed a positive correlation between CCL21 and Hsp90 levels (r > 0, (P < 0.05)). Binary logistic regression analysis suggested that high expression of CCL21 and Hsp90 were influencing factors for PE (OR >1, (P < 0.05)). The area under the receiver operating characteristic (AUC) curves of Hsp90 and CCL21 levels for predicting PE were 0.895 and 0.864, respectively, suggesting a good predictive value. Conclusion Serum CCL21 and Hsp90 show great potential as disease markers for PE prediction. Further trials are, however, required prior to clinical promotion.
Collapse
|
42
|
Extracellular vesicles for the treatment of preeclampsia. Tissue Cell 2022; 77:101860. [DOI: 10.1016/j.tice.2022.101860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/21/2022]
|
43
|
Kazemzadeh M, Martinez-Calderon M, Paek SY, Lowe M, Aguergaray C, Xu W, Chamley LW, Broderick NGR, Hisey CL. Classification of Preeclamptic Placental Extracellular Vesicles Using Femtosecond Laser Fabricated Nanoplasmonic Sensors. ACS Sens 2022; 7:1698-1711. [PMID: 35658424 DOI: 10.1021/acssensors.2c00378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Placental extracellular vesicles (EVs) play an essential role in pregnancy by protecting and transporting diverse biomolecules that aid in fetomaternal communication. However, in preeclampsia, they have also been implicated in contributing to disease progression. Despite their potential clinical value, current technologies cannot provide a rapid and effective means of differentiating between healthy and diseased placental EVs. To address this, a fabrication process called laser-induced nanostructuring of SERS-active thin films (LINST) was developed to produce scalable nanoplasmonic substrates that provide exceptional Raman signal enhancement and allow the biochemical fingerprinting of EVs. After validating the performance of LINST substrates with chemical standards, placental EVs from tissue explant cultures were characterized, demonstrating that preeclamptic and normotensive placental EVs have classifiably distinct Raman spectra following the application of advanced machine learning algorithms. Given the abundance of placental EVs in maternal circulation, these findings encourage immediate exploration of surface-enhanced Raman spectroscopy (SERS) of EVs as a promising method for preeclampsia liquid biopsies, while this novel fabrication process will provide a versatile and scalable substrate for many other SERS applications.
Collapse
Affiliation(s)
- Mohammadrahim Kazemzadeh
- Department of Mechanical and Mechatronics Engineering, University of Auckland, Auckland 1010, New Zealand.,Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9054, New Zealand
| | | | - Song Y Paek
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
| | - MoiMoi Lowe
- Department of Physics, University of Auckland, Auckland 1061, New Zealand
| | - Claude Aguergaray
- Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9054, New Zealand.,Department of Physics, University of Auckland, Auckland 1061, New Zealand
| | - Weiliang Xu
- Department of Mechanical and Mechatronics Engineering, University of Auckland, Auckland 1010, New Zealand.,Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9054, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand.,Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
| | - Neil G R Broderick
- Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9054, New Zealand.,Department of Physics, University of Auckland, Auckland 1061, New Zealand
| | - Colin L Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand.,Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand.,Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
44
|
The role of extracellular vesicles in endometrial receptivity and their potential in reproductive therapeutics and diagnosis. Reprod Biol 2022; 22:100645. [PMID: 35594727 DOI: 10.1016/j.repbio.2022.100645] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) are small, nanometre sized, membrane-enclosed structures released by cells and are thought to be crucial in cellular communication. The cargo of these vesicles includes lipids, proteins, RNAs and DNA, and control various biological processes in their target tissues depending on the parental and receiver cell's origin and phenotype. Recently data has accumulated in the role of EVs in embryo implantation and pregnancy, with EVs identified in the uterine cavity of women, sheep, cows, horses, and mice, in which they aid blastocyst and endometrial preparation for implantation. Herein is a critical review to decipher the role of extracellular vesicles in endometrial receptivity and their potential in reproductive therapies and diagnosis. The current knowledge of the function of embryo and endometrial derived EVs and their cargoes, with regards to their effect on implantation and receptivity are summarized and evaluated. The findings of the below review highlight that the combined knowledge on EVs deriving from the endometrium and embryo have the potential to be translated to various clinical applications including treatment, a diagnostic biomarker for diseases and a drug delivery tool to ultimately improve pregnancy rates.
Collapse
|
45
|
Rother N, Yanginlar C, Pieterse E, Hilbrands L, van der Vlag J. Microparticles in Autoimmunity: Cause or Consequence of Disease? Front Immunol 2022; 13:822995. [PMID: 35514984 PMCID: PMC9065258 DOI: 10.3389/fimmu.2022.822995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022] Open
Abstract
Microparticles (MPs) are small (100 nm - 1 um) extracellular vesicles derived from the plasma membrane of dying or activated cells. MPs are important mediators of intercellular communication, transporting proteins, nucleic acids and lipids from the parent cell to other cells. MPs resemble the state of their parent cells and are easily accessible when released into the blood or urine. MPs also play a role in the pathogenesis of different diseases and are considered as potential biomarkers. MP isolation and characterization is technically challenging and results in different studies are contradictory. Therefore, uniform guidelines to isolate and characterize MPs should be developed. Our understanding of MP biology and how MPs play a role in different pathological mechanisms has greatly advanced in recent years. MPs, especially if derived from apoptotic cells, possess strong immunogenic properties due to the presence of modified proteins and nucleic acids. MPs are often found in patients with autoimmune diseases where MPs for example play a role in the break of immunological tolerance and/or induction of inflammatory conditions. In this review, we describe the main techniques to isolate and characterize MPs, define the characteristics of MPs generated during cell death, illustrate different mechanism of intercellular communication via MPs and summarize the role of MPs in pathological mechanisms with a particular focus on autoimmune diseases.
Collapse
Affiliation(s)
- Nils Rother
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cansu Yanginlar
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Elmar Pieterse
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luuk Hilbrands
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
46
|
Ghafourian M, Mahdavi R, Akbari Jonoush Z, Sadeghi M, Ghadiri N, Farzaneh M, Mousavi Salehi A. The implications of exosomes in pregnancy: emerging as new diagnostic markers and therapeutics targets. Cell Commun Signal 2022; 20:51. [PMID: 35414084 PMCID: PMC9004059 DOI: 10.1186/s12964-022-00853-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vehicles (EVs) are a heterogeneous group of cell and membranous particles originating from different cell compartments. EVs participate in many essential physiological functions and mediate fetal-maternal communications. Exosomes are the smallest unit of EVs, which are delivered to the extracellular space. Exosomes can be released by the umbilical cord, placenta, amniotic fluid, and amniotic membranes and are involved in angiogenesis, endothelial cell migration, and embryo implantation. Also, various diseases such as gestational hypertension, gestational diabetes mellitus (GDM), preterm birth, and fetal growth restriction can be related to the content of placental exosomes during pregnancy. Due to exosomes' ability to transport signaling molecules and their effect on sperm function, they can also play a role in male and female infertility. In the new insight, exosomal miRNA can diagnose and treat infertilities disorders. In this review, we focused on the functions of exosomes during pregnancy. Video abstract.
Collapse
Affiliation(s)
- Mehri Ghafourian
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Roya Mahdavi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Akbari Jonoush
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahvash Sadeghi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nooshin Ghadiri
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Cellular and Molecular Research Center, Medical Basic Science Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Abdolah Mousavi Salehi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
47
|
Wang J, Xie X, Wu Y, Zhou Y, Li Q, Li Y, Xu X, Wang M, Murdiyarso L, Houck K, Hilton T, Chung D, Li M, Zhang JN, Dong J. Brain-Derived Extracellular Vesicles Induce Vasoconstriction and Reduce Cerebral Blood Flow in Mice. J Neurotrauma 2022; 39:879-890. [PMID: 35316073 DOI: 10.1089/neu.2021.0274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) impairs cerebrovascular autoregulation and reduces cerebral blood flow (CBF), leading to ischemic secondary injuries. We have shown that injured brains release brain-derived extracellular vesicles (BDEVs) into circulation, where they cause a systemic hypercoagulable state that rapidly turns into consumptive coagulopathy. BDEVs induce endothelial injury and permeability, leading to the hypothesis that they contribute to TBI-induced cerebrovascular dysregulation. In a study designed to test this hypothesis, we detected circulating BDEVs in C57BL/6J mice subjected to severe TBI, reaching peak levels of 3x104/µl at 3 hours post injury (71.2±21.5% of total annexin V-binding EVs). We further showed in an adaptive transfer model that 41.7±5.8% of non-injured mice died within 6 hours after being infused with 3x104/µl of BDEVs. BDEVs transmigrated through the vessel walls, induced rapid vasoconstriction by inducing calcium influx in vascular smooth muscle cells, and reduced CBF by 93.8±5.6% within 30 minutes after infusion. The CBF suppression was persistent in mice that eventually died but it recovered quickly in surviving mice. It was prevented by the calcium channel blocker nimodipine. When being separated, neither protein nor phospholipid components from the lethal number of BDEVs induced vasoconstriction, reduced CBF, and caused death. These results demonstrate a novel vasoconstrictive activity of BDEVs that depends on the structure of BDEVs and contributes to TBI-induced disseminated cerebral ischemia and sudden death.
Collapse
Affiliation(s)
- Jiwei Wang
- Tianjin Neurological Institute, 230967, Anshan road No.154, Tianjin, China, 300052;
| | - Xiaofeng Xie
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Yingang Wu
- University of Science and Technology of China, 12652, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine., Hefei, Anhui, China;
| | - Yuan Zhou
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Qifeng Li
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Ying Li
- Tianjin Neurological Institute, 230967, Tianjin, Tianjin, China;
| | - Xin Xu
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Min Wang
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | | | - Katie Houck
- Bloodworks Research institute, Seattle, United States;
| | | | - Dominic Chung
- Bloodworks Research institute, Seattle, United States;
| | - Min Li
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Jian-Ning Zhang
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Jingfei Dong
- Bloodworks Research Institute, Bloodworks Northwest, Seattle, Seattle, Washington, United States.,Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States;
| |
Collapse
|
48
|
Bakrania BA, George EM, Granger JP. Animal models of preeclampsia: investigating pathophysiology and therapeutic targets. Am J Obstet Gynecol 2022; 226:S973-S987. [PMID: 33722383 PMCID: PMC8141071 DOI: 10.1016/j.ajog.2020.10.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/01/2020] [Accepted: 10/19/2020] [Indexed: 02/03/2023]
Abstract
Animal models have been critical in investigating the pathogenesis, mediators, and even therapeutic options for a number of diseases, including preeclampsia. Preeclampsia is the leading cause of maternal and fetal morbidity and mortality worldwide. The placenta is thought to play a central role in the pathogenesis of this disease because it releases antiangiogenic and proinflammatory factors into the maternal circulation, resulting in the maternal syndrome. Despite the deleterious effects preeclampsia has been shown to have on the mother and baby during pregnancy and postpartum, there is still no effective treatment for this disease. Although clinical studies in patients are crucial to identify the involvement of pathogenic factors in preeclampsia, there are obvious limitations that prevent detailed investigation of the quantitative importance of time-dependent mechanisms involved in this syndrome. Animal models allow investigators to perform proof-of-concept studies and examine whether certain factors found in women with preeclampsia mediate hypertension and other manifestations of this disease. In this brief review, we summarize some of the more widely studied models used to investigate pathophysiological mechanisms that are thought to be involved in preeclampsia. These include models of placental ischemia, angiogenic imbalance, and maternal immune activation. Infusion of preeclampsia-related factors into animals has been widely studied to understand the specific mediators of this disease. These models have been included, in addition to a number of genetic models involved in overexpression of the renin-angiotensin system, complement activation, and trophoblast differentiation. Together, these models cover multiple mechanisms of preeclampsia from trophoblast dysfunction and impaired placental vascularization to the excess circulating placental factors and clinical manifestation of this disease. Most animal studies have been performed in rats and mice; however, we have also incorporated nonhuman primate models in this review. Preclinical animal models not only have been instrumental in understanding the pathophysiology of preeclampsia but also continue to be important tools in the search for novel therapeutic options for the treatment of this disease.
Collapse
Affiliation(s)
- Bhavisha A Bakrania
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Eric M George
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Joey P Granger
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS.
| |
Collapse
|
49
|
Brown PA. Differential and targeted vesiculation: pathologic cellular responses to elevated arterial pressure. Mol Cell Biochem 2022; 477:1023-1040. [PMID: 34989921 DOI: 10.1007/s11010-021-04351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles are small membrane-enclosed particles released during cell activation or injury. They have been investigated for several decades and found to be secreted in various diseases. Their pathogenic role is further supported by the presence of several important molecules among their cargo, including proteins, lipids, and nucleic acids. Many studies have reported enhanced and targeted extracellular vesicle biogenesis in diseases that involve chronic or transient elevation of arterial pressure resulting in endothelial dysfunction, within either the general circulatory system or specific local vascular beds. In addition, several associated pathologic processes have been studied and reported. However, the role of elevated pressure as a common pathogenic trigger across vascular domains and disease chronicity has not been previously described. This review will therefore summarize our current knowledge of the differential and targeted biogenesis of extracellular vesicles in major diseases that are characterized by elevated arterial pressure leading to endothelial dysfunction and propose a unified theory of pressure-induced extracellular vesicle-mediated pathogenesis.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Kingston 7, Jamaica.
| |
Collapse
|
50
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part II: Pathology: Part II: Pathology. J Extracell Vesicles 2022; 11:e12190. [PMID: 35041301 PMCID: PMC8765328 DOI: 10.1002/jev2.12190] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
It is clear from Part I of this series that extracellular vesicles (EVs) play a critical role in maintaining the homeostasis of most, if not all, normal physiological systems. However, the majority of our knowledge about EV signalling has come from studying them in disease. Indeed, EVs have consistently been associated with propagating disease pathophysiology. The analysis of EVs in biofluids, obtained in the clinic, has been an essential of the work to improve our understanding of their role in disease. However, to interfere with EV signalling for therapeutic gain, a more fundamental understanding of the mechanisms by which they contribute to pathogenic processes is required. Only by discovering how the EV populations in different biofluids change-size, number, and physicochemical composition-in clinical samples, may we then begin to unravel their functional roles in translational models in vitro and in vivo, which can then feedback to the clinic. In Part II of this review series, the functional role of EVs in pathology and disease will be discussed, with a focus on in vivo evidence and their potential to be used as both biomarkers and points of therapeutic intervention.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|