1
|
Valero P, Silva K, Valenzuela-Hinrichsen A, Vásquez A, Espinoza F, Lira F, Cornejo M, Fuentes G, González D, Moore-Carrasco R, van der Beek EM, Hillebrands JL, van Goor H, Grismaldo A, Sobrevia L. Shortcomings, limitations and gaps in physiological roles of extracellular vesicles in obesity. J Physiol 2024. [PMID: 39470472 DOI: 10.1113/jp286955] [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: 05/17/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in mediating communication between cells across species and kingdoms. The intercellular communication facilitated by EVs through autocrine and paracrine signalling mechanisms is essential for cell survival, maintaining normal metabolic functions and ensuring overall bodily homeostasis and health. Extracellular vesicles are present in various bodily fluids, such as pleural effusions, plasma, breast milk, amniotic fluid, semen and saliva. Additionally, the generation and release of EVs contribute to the removal of cellular waste. Patients with obesity exhibit a higher release and amount of circulating EVs than individuals with normal weight. This increased EV release in obesity might contribute to the inflammatory state characteristic of this metabolic condition, because higher levels of pro-inflammatory molecules are found within their cargo. However, interpreting results related to EV abundance, cargo and biological actions can be complicated by several factors; these include variations in cell sources, a wide age range (from children to the elderly), a mix of females and males, medication use and health status, a range of body weights (from normal weight to morbid obesity) and differences between in vitro assays using cell lines versus primary cultures. This article addresses the shortcomings, limitations and gaps in knowledge, providing a framework for enhancing our understanding of the physiological effects of EVs on obesity.
Collapse
Affiliation(s)
- Paola Valero
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Katherin Silva
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Andrés Valenzuela-Hinrichsen
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Vásquez
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Espinoza
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Lira
- Faculty of Medicine, Universidad de Antofagasta, Antofagasta, Chile
| | - Marcelo Cornejo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta, Chile
| | - Gonzalo Fuentes
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Daniel González
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | | | - Eline M van der Beek
- Department of Pediatrics, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- Nestlé Institute for Health Sciences, Nestlé Research, Societé des Produits de Nestlé, Lausanne, Switzerland
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Adriana Grismaldo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
| | - Luis Sobrevia
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
2
|
Xing Y, Kang L, Chen L, Li Y, Lu D. Research progress of exosomes in pathogenesis and treatment of preeclampsia. J Obstet Gynaecol Res 2024. [PMID: 39434205 DOI: 10.1111/jog.16106] [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: 07/02/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024]
Abstract
AIM Preeclampsia (PE) is a critical and severe disease in obstetrics, which seriously affects maternal and neonatal life safety and long-term prognosis. However, the etiology and pathogenesis of PE are complex, and no unified conclusion has been reached. The types and number of exosomes and their transport substances in PE patients changed. The study of exosomes in PE patients helps clarify the etiology, diagnosis, effective treatment, accurate monitoring, and prognosis. METHOD The published articles were reviewed. RESULTS Exosomes may affect endothelial and vascular production and function, participate in maternal-fetal immune regulation, and transport substances such as miRNAs, lncRNAs, and proteins involved in the development of PE. Detection of the contents of exosomes can help in the early diagnosis of PE, and can help to improve PE by inhibiting the action of exosomes or preventing their binding to target organs. CONCLUSION Exosomes may be involved in the development of PE, and exosomes can be used as markers for predicting the onset of PE and tracking the disease process and determining the prognosis, and exosomes have great potential in the treatment of PE.
Collapse
Affiliation(s)
- Yue Xing
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Luyao Kang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Lu Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Youyou Li
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Dan Lu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
3
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
4
|
Xu X, Liu S, Gao Y, Cheng L. Epigallocatechin gallate (EGCG) alleviates inflammation and endothelial dysfunction and improves pregnancy outcomes in preeclampsia (PE)-like rats via eNOS/Nrf2/HO-1 pathway. J Reprod Immunol 2024; 164:104263. [PMID: 38838579 DOI: 10.1016/j.jri.2024.104263] [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: 03/03/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND AND PURPOSE Epigallocatechin gallate (EGCG), a natural antioxidant, has shown protective effect in many diseases. We explore the effect and potential regulatory mechanisms of EGCG in preeclampsia (PE)-like rats. METHODS AND MATERIALS PE was mimicked in pregnant rats. EGCG was orally administered at a dosage of 25(Low, L) or 50 mg/kg (High, H) from gestational day (GD) 6-17. The blood pressure signatures, heart rates were monitored. The 24-h proteinuria and serum were analyzed. On GD 18, rats were sacrificed, and pups and placentas were weighed. Kidneys and placentas were analyzed using immunohistochemistry (IHC) and hematoxylin-eosin staining (H&E). Placentas were examined using western blot for sFlt1, eNOS, Nrf2, HO-1, SLC7A11. MDA, GSH, GPx and Fe2+ were measured. RESULTS EGCG inhibits systolic blood pressure, BUN, CREA, ALT, AST, UA and proteinuria levels in PE-like rats. EGCG enhances the pup weight and crown-rump length and reduces the rate of fetus growth restriction in PE group. Endothelial dysfunction and infiltration of inflammatory cells were found in kidney cortex and placenta tissues in PE group and were inhibited by EGCG treatment. sFlt1 was activated in placentas in PE group and inhibited by EGCG while eNOS/Nrf2/HO-1 were inhibited in PE group and restored by EGCG. MDA and Fe concentrations were elevated in PE group and reduced by EGCG while the GSH level, SLC7A11 and the GPx activity were inhibited in PE group and restored by EGCG. CONCLUSION EGCG alleviates inflammation, endothelial dysfunction and placental ferroptosis, improves pregnancy outcomes in PE-like rats via eNOS/Nrf2/HO-1.
Collapse
Affiliation(s)
- Xinran Xu
- Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Shasha Liu
- Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Yiping Gao
- Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Lan Cheng
- Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China.
| |
Collapse
|
5
|
Torres-Torres J, Espino-y-Sosa S, Martinez-Portilla R, Borboa-Olivares H, Estrada-Gutierrez G, Acevedo-Gallegos S, Ruiz-Ramirez E, Velasco-Espin M, Cerda-Flores P, Ramirez-Gonzalez A, Rojas-Zepeda L. A Narrative Review on the Pathophysiology of Preeclampsia. Int J Mol Sci 2024; 25:7569. [PMID: 39062815 PMCID: PMC11277207 DOI: 10.3390/ijms25147569] [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: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Preeclampsia (PE) is a multifactorial pregnancy disorder characterized by hypertension and proteinuria, posing significant risks to both maternal and fetal health. Despite extensive research, its complex pathophysiology remains incompletely understood. This narrative review aims to elucidate the intricate mechanisms contributing to PE, focusing on abnormal placentation, maternal systemic response, oxidative stress, inflammation, and genetic and epigenetic factors. This review synthesizes findings from recent studies, clinical trials, and meta-analyses, highlighting key molecular and cellular pathways involved in PE. The review integrates data on oxidative stress biomarkers, angiogenic factors, immune interactions, and mitochondrial dysfunction. PE is initiated by poor placentation due to inadequate trophoblast invasion and improper spiral artery remodeling, leading to placental hypoxia. This triggers the release of anti-angiogenic factors such as soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), causing widespread endothelial dysfunction and systemic inflammation. Oxidative stress, mitochondrial abnormalities, and immune dysregulation further exacerbate the condition. Genetic and epigenetic modifications, including polymorphisms in the Fms-like tyrosine kinase 1 (FLT1) gene and altered microRNA (miRNA) expression, play critical roles. Emerging therapeutic strategies targeting oxidative stress, inflammation, angiogenesis, and specific molecular pathways like the heme oxygenase-1/carbon monoxide (HO-1/CO) and cystathionine gamma-lyase/hydrogen sulfide (CSE/H2S) pathways show promise in mitigating preeclampsia's effects. PE is a complex disorder with multifactorial origins involving abnormal placentation, endothelial dysfunction, systemic inflammation, and oxidative stress. Despite advances in understanding its pathophysiology, effective prevention and treatment strategies remain limited. Continued research is essential to develop targeted therapies that can improve outcomes for both mothers and their babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Salvador Espino-y-Sosa
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Raigam Martinez-Portilla
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Hector Borboa-Olivares
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Guadalupe Estrada-Gutierrez
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Sandra Acevedo-Gallegos
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Erika Ruiz-Ramirez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Martha Velasco-Espin
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Pablo Cerda-Flores
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Andrea Ramirez-Gonzalez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Medicine Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| |
Collapse
|
6
|
Countouris ME, Catov JM, Zhu J, de Jong N, Brands J, Chen X, Parks WT, Berlacher KL, Gandley RE, Straub AC, Villanueva FS. Association of Hypertensive Disorders of Pregnancy With Coronary Microvascular Dysfunction 8 to 10 Years After Delivery. Circ Cardiovasc Imaging 2024; 17:e016561. [PMID: 38771901 PMCID: PMC11115371 DOI: 10.1161/circimaging.124.016561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/17/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDP) are associated with subsequent adverse cardiac remodeling and cardiovascular disease. The role of myocardial microvascular disease among individuals with HDP and left ventricular (LV) remodeling as a potential link to cardiovascular disease is unknown. We aimed to determine whether individuals with HDP history have coronary microvascular dysfunction measured by coronary flow reserve 8 to 10 years after delivery and whether microvascular dysfunction correlates with LV remodeling. METHODS Individuals with pregnancies delivered from 2008 to 2010 underwent burst-replenishment myocardial contrast echocardiography (2017-2020) to quantify myocardial perfusion at rest and during dobutamine stress. Video intensity versus time data were used to derive β, the rate of rise of video intensity, a correlate for myocardial blood flow. Coronary flow reserve was calculated as the ratio of β at peak stress to β at rest, averaged across LV myocardial regions of interest. RESULTS We studied 91 individuals (aged 38±6 and 9.1±0.9 years postdelivery) and 19 with a history of HDP. Individuals with coronary microvascular dysfunction (coronary flow reserve <2.0; n=13) had a higher proportion of HDP (46.2% versus 16.7%; P=0.026) and higher prepregnancy body mass index, baseline heart rate, and hemoglobin A1c compared with those without microvascular dysfunction. The association of coronary flow reserve and HDP was attenuated after adjusting for cardiometabolic factors (P=0.133). In exploratory subgroup analyses, individuals with both LV remodeling (relative wall thickness >0.42) and HDP (n=12) had the highest proportion of microvascular dysfunction (41.7% versus +HDP-LV remodeling [n=7] 14.3%; -HDP+LV remodeling [n=26] 7.7%; P=0.0498). CONCLUSIONS In this small study, HDP history is associated with coronary microvascular dysfunction 1 decade after delivery, findings that may, in part, be driven by metabolic factors including obesity and diabetes. Microvascular dysfunction may contribute to cardiovascular disease among individuals with a history of HDP.
Collapse
Affiliation(s)
- Malamo E Countouris
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
| | - Janet M Catov
- Department of Obstetrics, Gynecology, and Reproductive Sciences (J.M.C., R.E.G.), University of Pittsburgh, PA
- Department of Epidemiology (J.M.C.), University of Pittsburgh, PA
| | - Jianhui Zhu
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
| | - Nikki de Jong
- Division of Cardiology, Erasmus Medical Center, Rotterdam, the Netherlands (N.d.J.)
| | - Judith Brands
- Department of Library, Information and Communication Technologies Services and Archive, Enschede, the Netherlands (J.B.)
| | - Xucai Chen
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
| | - W Tony Parks
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada (W.T.P.)
| | - Kathryn L Berlacher
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
| | - Robin E Gandley
- Department of Obstetrics, Gynecology, and Reproductive Sciences (J.M.C., R.E.G.), University of Pittsburgh, PA
| | - Adam C Straub
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
- Department of Pharmacology and Chemical Biology (A.C.S.), University of Pittsburgh, PA
| | - Flordeliza S Villanueva
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, PA (M.E.C., J.Z., X.C., K.L.B., A.C.S., F.S.V.)
| |
Collapse
|
7
|
Nishi K, Modi D. Placental exosomes in pregnancy and preeclampsia. Am J Reprod Immunol 2024; 91:e13857. [PMID: 38716824 DOI: 10.1111/aji.13857] [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: 01/02/2024] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/24/2024] Open
Abstract
Preeclampsia, poses significant risks to both maternal and fetal well-being. Exosomes released by the placenta play a crucial role in intercellular communication and are recognized as potential carriers of essential information for placental development. These exosomes transport a payload of proteins, nucleic acids, and lipids that mirror the placental microenvironment. This review delves into the functional roles of placental exosomes and its contents shedding light on their involvement in vascular regulation and immune modulation in normal pregnancy. Discernible changes are reported in the composition and quantity of placental exosome contents in pregnancies affected by preeclampsia. The exosomes from preeclamptic mothers affect vascularization and fetal kidney development. The discussion also explores the implications of utilizing placental exosomes as biomarkers and the prospects of translating these findings into clinical applications. In conclusion, placental exosomes hold promise as a valuable avenue for deciphering the complexities of preeclampsia, providing crucial diagnostic and prognostic insights. As the field progresses, a more profound comprehension of the distinct molecular signatures carried by placental exosomes may open doors to innovative strategies for managing and offering personalized care to pregnancies affected by preeclampsia.
Collapse
Affiliation(s)
- Kumari Nishi
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
8
|
Cooke WR, Jiang P, Ji L, Bai J, Jones GD, Lo YMD, Redman C, Vatish M. Differential 5'-tRNA Fragment Expression in Circulating Preeclampsia Syncytiotrophoblast Vesicles Drives Macrophage Inflammation. Hypertension 2024; 81:876-886. [PMID: 38362745 PMCID: PMC10956686 DOI: 10.1161/hypertensionaha.123.22292] [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: 10/25/2023] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND The relationship between placental pathology and the maternal syndrome of preeclampsia is incompletely characterized. Mismatch between placental nutrient supply and fetal demands induces stress in the syncytiotrophoblast, the layer of placenta in direct contact with maternal blood. Such stress alters the content and increases the release of syncytiotrophoblast extracellular vesicles (STB-EVs) into the maternal circulation. We have previously shown 5'-tRNA fragments (5'-tRFs) constitute the majority of small RNA in STB-EVs in healthy pregnancy. 5'-tRFs are produced in response to stress. We hypothesized STB-EV 5'-tRF release might change in preeclampsia. METHODS We perfused placentas from 8 women with early-onset preeclampsia and 6 controls, comparing small RNA expression in STB-EVs. We used membrane-affinity columns to isolate maternal plasma vesicles and investigate placental 5'-tRFs in vivo. We quantified 5'-tRFs from circulating STB-EVs using a placental alkaline phosphatase immunoassay. 5'-tRFs and scrambled RNA controls were added to monocyte, macrophage and endothelial cells in culture to investigate transcriptional responses. RESULTS 5'-tRFs constitute the majority of small RNA in STB-EVs from both preeclampsia and normal pregnancies. More than 900 small RNA fragments are differentially expressed in preeclampsia STB-EVs. Preeclampsia-dysregulated 5'-tRFs are detectable in maternal plasma, where we identified a placentally derived load. 5'-tRF-Glu-CTC, the most abundant preeclampsia-upregulated 5'-tRF in perfusion STB-EVs, is also increased in preeclampsia STB-EVs from maternal plasma. 5'-tRF-Glu-CTC induced inflammation in macrophages but not monocytes. The conditioned media from 5'-tRF-Glu-CTC-activated macrophages reduced eNOS (endothelial NO synthase) expression in endothelial cells. CONCLUSIONS Increased release of syncytiotrophoblast-derived vesicle-bound 5'-tRF-Glu-CTC contributes to preeclampsia pathophysiology.
Collapse
Affiliation(s)
- William R Cooke
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Peiyong Jiang
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Lu Ji
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Jinyue Bai
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Gabriel Davis Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Y M Dennis Lo
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Christopher Redman
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| |
Collapse
|
9
|
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
|
10
|
Weihrauch D, Keszler A, Broeckel G, Aranda E, Lindemer B, Lohr NL. Red light mediates the exocytosis of vasodilatory vesicles from cultured endothelial cells: a cellular, and ex vivo murine model. Photochem Photobiol Sci 2024; 23:355-364. [PMID: 38277065 PMCID: PMC10917865 DOI: 10.1007/s43630-023-00522-1] [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: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024]
Abstract
We have previously established that 670 nm energy induces relaxation of blood vessels via an endothelium derived S-nitrosothiol (RSNO) suggested to be embedded in vesicles. Here, we confirm that red light facilitates the exocytosis of this vasodilator from cultured endothelial cells and increases ex vivo blood vessel diameter. Ex vivo pressurized and pre-constricted facial arteries from C57Bl6/J mice relaxed 14.7% of maximum diameter when immersed in the medium removed from red-light exposed Bovine Aortic Endothelial Cells. In parallel experiments, 0.49 nM RSNO equivalent species was measured in the medium over the irradiated cells vs dark control. Electron microscopy of light exposed endothelium revealed significant increases in the size of the Multi Vesicular Body (MVB), a regulator of exosome trafficking, while RSNO accumulated in the MVBs as detected with immunogold labeling electron microscopy (1.8-fold of control). Moreover, red light enhanced the presence of F-actin related stress fibers (necessary for exocytosis), and the endothelial specific marker VE-cadherin levels suggesting an endothelial origin of the extracellular vesicles. Flow cytometry coupled with DAF staining, an indirect sensor of nitric oxide (NO), indicated significant amounts of NO within the extracellular vesicles (1.4-fold increase relative to dark control). Therefore, we further define the mechanism on the 670 nm light mediated traffic of endothelial vasodilatory vesicles and plan to leverage this insight into the delivery of red-light therapies.
Collapse
Affiliation(s)
- Dorothee Weihrauch
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Anesthesiology, Milwaukee, WI, USA
- Department of Plastic Surgery, Milwaukee, WI, USA
| | - Agnes Keszler
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Grant Broeckel
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva Aranda
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Anesthesiology, Milwaukee, WI, USA
| | - Brian Lindemer
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nicole L Lohr
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Clement J Zablocki VA Medical Center, Milwaukee, WI, USA.
- Cardiovascular Institute, University of Birmingham, Alabama, USA.
| |
Collapse
|
11
|
Neves KB, Rios FJ, Sevilla‐Montero J, Montezano AC, Touyz RM. Exosomes and the cardiovascular system: role in cardiovascular health and disease. J Physiol 2023; 601:4923-4936. [PMID: 35306667 PMCID: PMC10953460 DOI: 10.1113/jp282054] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/15/2022] [Indexed: 11/16/2023] Open
Abstract
Exosomes, which are membrane-bound extracellular vesicles (EVs), are generated in the endosomal compartment of almost all eukaryotic cells. They are formed upon the fusion of multivesicular bodies and the plasma membrane and carry proteins, nucleic acids, lipids and other cellular constituents from their parent cells. Multiple factors influence their production including cell stress and injury, humoral factors, circulating toxins, and oxidative stress. They play an important role in intercellular communication, through their ability to transfer their cargo (proteins, lipids, RNAs) from one cell to another. Exosomes have been implicated in the pathophysiology of various diseases including cardiovascular disease (CVD), cancer, kidney disease, and inflammatory conditions. In addition, circulating exosomes may act as biomarkers for diagnostic and prognostic strategies for several pathological processes. In particular exosome-containing miRNAs have been suggested as biomarkers for the diagnosis and prognosis of myocardial injury, stroke and endothelial dysfunction. They may also have therapeutic potential, acting as vectors to deliver therapies in a targeted manner, such as the delivery of protective miRNAs. Transfection techniques are in development to load exosomes with desired cargo, such as proteins or miRNAs, to achieve up-regulation in the host cell or tissue. These advances in the field have the potential to assist in the detection and monitoring progress of a disease in patients during its early clinical stages, as well as targeted drug delivery.
Collapse
Affiliation(s)
- Karla B. Neves
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowUK
| | - Francisco J. Rios
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowUK
| | - Javier Sevilla‐Montero
- Biomedical Research Institute La Princesa Hospital (IIS‐IP)Department of MedicineSchool of MedicineUniversidad Autónoma of Madrid (UAM)MadridSpain
| | | | - Rhian M. Touyz
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowUK
- Research Institute of the McGill University Health Centre (RI‐MUHC)McGill UniversityMontrealCanada
| |
Collapse
|
12
|
Kaihara JNS, Minami CK, Peraçoli MTS, Romão-Veiga M, Ribeiro-Vasques VR, Peraçoli JC, Palei ACT, Cavalli RC, Nunes PR, Luizon MR, Sandrim VC. Plasma eNOS Concentration in Healthy Pregnancy and in Hypertensive Disorders of Pregnancy: Evidence of Reduced Concentrations in Pre-Eclampsia from Two Independent Studies. Diseases 2023; 11:155. [PMID: 37987266 PMCID: PMC10660730 DOI: 10.3390/diseases11040155] [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: 09/02/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
Hypertensive disorders of pregnancy (HDP), comprising gestational hypertension (GH) and pre-eclampsia (PE), are leading causes of maternal and perinatal morbidity and mortality. Both GH and PE are characterized by new-onset hypertension, but PE additionally includes proteinuria and/or end-organ damage. Impaired nitric oxide (NO) bioavailability may lead to endothelial dysfunction in GH and PE, and the primary source of vascular NO is endothelial NO synthase (eNOS). However, no previous study has investigated plasma eNOS concentrations in patients with GH and PE. In this study, we compared plasma eNOS concentrations in healthy pregnancies and HDP in two independent cohorts. The primary study included 417 subjects, with 43 non-pregnant (NP) and 156 healthy pregnant (HP) women and 122 patients with GH and 96 with PE. The replication study included 85 pregnant women (41 healthy and 44 pre-eclamptic). Plasma concentrations of eNOS were measured using a commercial ELISA kit provided by R&D Systems, and plasma nitrite concentrations were assessed using two ozone-based chemiluminescence assays. Correlations between plasma eNOS concentrations and plasma nitrite concentrations, as well as clinical and biochemical parameters, were evaluated by either Spearman's or Pearson's tests. In the primary study, NP women and HDP had significantly lower plasma eNOS concentrations compared to HP; concentrations were even lower in PE compared to GH. Plasma eNOS concentrations were reduced but not significant in early-onset PE, PE with severe features, preterm birth, and intrauterine growth restriction. No correlation was observed between plasma eNOS and nitrite levels. In HDP, there was a significant positive correlation between levels of eNOS and hemoglobin (r = 0.1496, p = 0.0336) as well as newborn weight (r = 0.1487, p = 0.0316). Conversely, a negative correlation between eNOS levels and proteinuria was observed (r = -0.2167, p = 0.0179). The replication study confirmed significantly reduced plasma concentrations of eNOS in PE compared to HP. Our findings provide evidence of reduced plasma eNOS concentrations in HDP; they were particularly lower in PE compared to GH and HP in two independent studies.
Collapse
Affiliation(s)
- Julyane N. S. Kaihara
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (J.N.S.K.); (C.K.M.); (P.R.N.); (M.R.L.)
| | - Caroline K. Minami
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (J.N.S.K.); (C.K.M.); (P.R.N.); (M.R.L.)
| | - Maria T. S. Peraçoli
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.T.S.P.); (M.R.-V.); (J.C.P.)
| | - Mariana Romão-Veiga
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.T.S.P.); (M.R.-V.); (J.C.P.)
| | - Vanessa R. Ribeiro-Vasques
- Department of Chemistry and Biological Sciences, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil;
| | - José C. Peraçoli
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.T.S.P.); (M.R.-V.); (J.C.P.)
| | - Ana C. T. Palei
- Department of Surgery, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Ricardo C. Cavalli
- Department of Gynecology and Obstetrics, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto 14049-900, SP, Brazil;
| | - Priscila R. Nunes
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (J.N.S.K.); (C.K.M.); (P.R.N.); (M.R.L.)
| | - Marcelo R. Luizon
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (J.N.S.K.); (C.K.M.); (P.R.N.); (M.R.L.)
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Valeria C. Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (J.N.S.K.); (C.K.M.); (P.R.N.); (M.R.L.)
| |
Collapse
|
13
|
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: 4] [Impact Index Per Article: 4.0] [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
|
14
|
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
|
15
|
Jia L, Huang X, Peng H, Jia Y, Zhang R, Wei Y, Wei M, Wang R, Li H, He Q, Wang K. Pregnancy-specific beta-1-glycoprotein 1-enriched exosomes are involved in the regulation of vascular endothelial cell function during pregnancy. Placenta 2023; 139:138-147. [PMID: 37392715 DOI: 10.1016/j.placenta.2023.06.014] [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: 03/03/2023] [Revised: 05/29/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023]
Abstract
INTRODUCTION Pregnancy is a dynamic time period associated with significant physiological changes in the cardiovascular system. It is well known that during pregnancy, the placenta secretes a variety of molecular signals, including exosomes, into the maternal circulation to adapt to increased blood volume and maintain blood pressure at normotensive levels. METHODS In the present study, we compared the effects of exosomes derived from the peripheral blood serum of nonpregnant women (NP-Exo) and pregnant women with uncomplicated pregnancy (P-Exo) on endothelial cell function. We also analyzed the proteomic profiles of these two groups of exosomes and the molecular mechanisms underlying the effect of exosome cargoes on vascular endothelial cell function. RESULTS We found that P-Exo were positively involved in regulating the function of human umbilical vein endothelial cell (HUVEC) and promoting the release of nitric oxide (NO). Furthermore, we revealed that trophoblast-derived pregnancy-specific beta-1-glycoprotein 1 (PSG1)-enriched exosomes treatment induced the promotion of HUVEC proliferation and migration as well as the release of NO. In addition, we found that P-Exo maintained blood pressure at normal levels in mice. DISCUSSION These results suggested that PSG1-enriched exosomes derived from maternal peripheral blood regulate the function of vascular endothelial cells and play an important role in maintaining maternal blood pressure during pregnancy.
Collapse
Affiliation(s)
- Linyan Jia
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaojie Huang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Peng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanhui Jia
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruonan Zhang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mengtian Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruixue Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Li
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qizhi He
- Department of Pathology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
16
|
Espinosa CA, Khan W, Khanam R, Das S, Khalid J, Pervin J, Kasaro MP, Contrepois K, Chang AL, Phongpreecha T, Michael B, Ellenberger M, Mehmood U, Hotwani A, Nizar A, Kabir F, Wong RJ, Becker M, Berson E, Culos A, De Francesco D, Mataraso S, Ravindra N, Thuraiappah M, Xenochristou M, Stelzer IA, Marić I, Dutta A, Raqib R, Ahmed S, Rahman S, Hasan ASMT, Ali SM, Juma MH, Rahman M, Aktar S, Deb S, Price JT, Wise PH, Winn VD, Druzin ML, Gibbs RS, Darmstadt GL, Murray JC, Stringer JSA, Gaudilliere B, Snyder MP, Angst MS, Rahman A, Baqui AH, Jehan F, Nisar MI, Vwalika B, Sazawal S, Shaw GM, Stevenson DK, Aghaeepour N. Multiomic signals associated with maternal epidemiological factors contributing to preterm birth in low- and middle-income countries. SCIENCE ADVANCES 2023; 9:eade7692. [PMID: 37224249 PMCID: PMC10208584 DOI: 10.1126/sciadv.ade7692] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
Preterm birth (PTB) is the leading cause of death in children under five, yet comprehensive studies are hindered by its multiple complex etiologies. Epidemiological associations between PTB and maternal characteristics have been previously described. This work used multiomic profiling and multivariate modeling to investigate the biological signatures of these characteristics. Maternal covariates were collected during pregnancy from 13,841 pregnant women across five sites. Plasma samples from 231 participants were analyzed to generate proteomic, metabolomic, and lipidomic datasets. Machine learning models showed robust performance for the prediction of PTB (AUROC = 0.70), time-to-delivery (r = 0.65), maternal age (r = 0.59), gravidity (r = 0.56), and BMI (r = 0.81). Time-to-delivery biological correlates included fetal-associated proteins (e.g., ALPP, AFP, and PGF) and immune proteins (e.g., PD-L1, CCL28, and LIFR). Maternal age negatively correlated with collagen COL9A1, gravidity with endothelial NOS and inflammatory chemokine CXCL13, and BMI with leptin and structural protein FABP4. These results provide an integrated view of epidemiological factors associated with PTB and identify biological signatures of clinical covariates affecting this disease.
Collapse
Affiliation(s)
- Camilo A. Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sayan Das
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Javairia Khalid
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Margaret P. Kasaro
- University of North Carolina Global Projects Zambia, Lusaka, Zambia
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanaphong Phongpreecha
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ambreen Nizar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Neal Ravindra
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Melan Thuraiappah
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Arup Dutta
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Rubhana Raqib
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | | | | | - Said M. Ali
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Mohamed H. Juma
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Monjur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Shaki Aktar
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Saikat Deb
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Joan T. Price
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Paul H. Wise
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurice L. Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald S. Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary L. Darmstadt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeffrey S. A. Stringer
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anisur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Abdullah H. Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fyezah Jehan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Bellington Vwalika
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Sunil Sazawal
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Huang X, Jia L, Jia Y, Xu X, Wang R, Wei M, Li H, Peng H, Wei Y, He Q, Wang K. sFlt-1-enriched exosomes induced endothelial cell dysfunction and a preeclampsia-like phenotype in mice. Cytokine 2023; 166:156190. [PMID: 37062152 DOI: 10.1016/j.cyto.2023.156190] [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: 11/11/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/18/2023]
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy characterized by maternal endothelial dysfunction and end-organ damage. Our previous work demonstrated that PE patient-derived exosomes contained higher levels of soluble FMS-like tyrosine kinase-1 (sFlt-1) and significantly induced endothelial dysfunction and PE development. However, the mechanisms underlying the effect of sFlt-1-enriched exosomes (sFlt-1-Exo) on PE development are poorly characterized. Here, we revealed that trophoblast-derived sFlt-1-Exo treatment induced significant inhibition of human umbilical vein endothelial cell (HUVEC) migration and tube formation, as well as an increase in sFlt-1 secretion. Mechanistically, we found that the increased sFlt-1 secretion in the cell culture medium was attributed to enhanced transcription of sFlt-1 in HUVECs. Importantly, we observed that treating pregnant mice with sFlt-1-Exo or recombinant mouse sFlt-1 triggered a preeclampsia-like phenotype, characterized by elevated blood pressure, proteinuria, increased plasma sFlt-1 and adverse pregnancy outcomes. These results strongly suggested that sFlt-1-Exo-induced endothelial dysfunction could be partially attributed to the upregulation of sFlt-1 in endothelial cells, potentially leading to the development of a preeclampsia-like phenotype in mice.
Collapse
Affiliation(s)
- Xiaojie Huang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Linyan Jia
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanhui Jia
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xianghong Xu
- Department of Biobank, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruixue Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mengtian Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Li
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Peng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
19
|
Myocardial perfusion and function dichotomy in growth restricted preterm infants. J Dev Orig Health Dis 2023; 14:302-310. [PMID: 36408644 DOI: 10.1017/s2040174422000630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Compared to preterm appropriate for gestational age (AGA) fetuses, fetuses with fetal growth restriction (FGR) have earlier visualisation of coronary artery blood flow (CABF) but impaired cardiac function. This dichotomy remains uncharacterised during postnatal life. This study compared CABF and cardiac function in preterm FGR infants, against AGA infants during the postnatal period. FGR was defined as birthweight < 10th centile for gestation and sex with absent/reversed antenatal umbilical artery Doppler. Diastolic CABF was measured in the left anterior descending coronary artery. Twenty-eight FGR infants were compared with 26 AGA infants (gestation and birthweight, 29.7 ± 1.3 vs 29.9 ± 1 weeks, P = 0.6 and 918 ± 174 vs 1398 ± 263g, P < 0.001, respectively). Echocardiography was performed in the second week of life. FGR infants had higher CABF (velocity time integral, 2.4 ± 0.9 vs 1.6 ± 0.8 cm, P = 0.002). Diastolic function was impaired (↑ trans-mitral E/A ratio in FGR infants; 0.84 ± 0.05 vs 0.79 ± 0.03, P = 0.0002) while the systolic function was also affected (mean velocity of circumferential fibre shortening [mVCFc], 1.9 ± 0.3 vs 2.7 ± 0.5 circ/s, P < 0.001). Indexing CABF to cardiac function noted significant differences between the groups (CABF: E/A [FGR vs AGA], 2.9 ± 1.1 vs 2.1 ± 1, P = 0.01 and CABF: mVCFc [FGR vs AGA], 1.3 ± 0.5 vs 0.6 ± 0.3, P < 0.001). Diastolic blood pressure (BP) was significantly higher, and CABF to diastolic BP ratio trended higher in FGR infants (30 ± 2 vs 25 ± 3 mmHg, P < 0.001 and 0.08 ± 0.03 vs 0.06 ± 0.03, P = 0.059, respectively). Greater CABF in FGR infants did not translate into better cardiac function. This dichotomy may be a persistent response to fetal hypoxaemia (fetal programming) and/or reflection of altered cardiac architecture.
Collapse
|
20
|
Gonzalez Fernandez J, Moncayo Arlandi J, Ochando A, Simon C, Vilella F. The role of extracellular vesicles in intercellular communication in human reproduction. Clin Sci (Lond) 2023; 137:281-301. [PMID: 36762584 DOI: 10.1042/cs20220793] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Embryo-maternal cross-talk has emerged as a vitally important process for embryo development and implantation, which is driven by secreted factors and extracellular vesicles (EVs). The EV cargo of bioactive molecules significantly influences target cells and primes them for critical stages of reproductive biology, including embryo development, adhesion, and implantation. Recent research has suggested that EVs and their cargo represent a powerful non-invasive tool that can be leveraged to assess embryo and maternal tissue quality during assisted reproduction treatments. Here, we review the current scientific literature regarding the intercellular cross-talk between embryos and maternal tissues from fertilization to implantation, focusing on human biology and signaling mechanisms identified in animal models.
Collapse
Affiliation(s)
- Javier Gonzalez Fernandez
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Javier Moncayo Arlandi
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Ana Ochando
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Carlos Simon
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Felipe Vilella
- Carlos Simon Foundation, INCLIVA Health Research Institute, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| |
Collapse
|
21
|
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: 3.0] [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
|
22
|
Dela Justina V, Dos Passos Júnior RR, Lima VV, Giachini FR. Evidence of Nitric Oxide Impairment During Hypertensive Pregnancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:99-125. [PMID: 37466771 DOI: 10.1007/978-3-031-32554-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Hypertensive disorders of pregnancy complicate up to 10% of pregnancies worldwide, and they can be classified into (1) gestational hypertension, (2) preeclampsia, (3) chronic hypertension and (4) chronic hypertension with preeclampsia. Nitric oxide (NO) plays an essential role in the haemodynamic adaptations observed during pregnancy. It has been shown that the nitric oxide pathway's dysfunction during pregnancy is associated with placental- and vascular-related diseases such as hypertensive disorders of pregnancy. This review aims to present a brief definition of hypertensive disorders of pregnancy and physiological maternal cardiovascular adaptations during pregnancy. We also detail how NO signalling is altered in the (a) systemic vasculature, (b) uterine artery/spiral arteries, (c) implantation and (d) placenta of hypertensive disorders during pregnancy. We conclude by summarizing the anti-hypertensive therapy of hypertensive disorders of pregnancy as a specific management strategy.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos Júnior
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Victor Vitorino Lima
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Fernanda Regina Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| |
Collapse
|
23
|
Preeclampsia and syncytiotrophoblast membrane extracellular vesicles (STB-EVs). Clin Sci (Lond) 2022; 136:1793-1807. [PMID: 36511102 DOI: 10.1042/cs20220149] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 12/15/2022]
Abstract
Preeclampsia (PE) is a hypertensive complication of pregnancy that affects 2-8% of women worldwide and is one of the leading causes of maternal deaths and premature birth. PE can occur early in pregnancy (<34 weeks gestation) or late in pregnancy (>34 weeks gestation). Whilst the placenta is clearly implicated in early onset PE (EOPE), late onset PE (LOPE) is less clear with some believing the disease is entirely maternal whilst others believe that there is an interplay between maternal systems and the placenta. In both types of PE, the syncytiotrophoblast (STB), the layer of the placenta in direct contact with maternal blood, is stressed. In EOPE, the STB is oxidatively stressed in early pregnancy (leading to PE later in gestation- the two-stage model) whilst in LOPE the STB is stressed because of villous overcrowding and senescence later in pregnancy. It is this stress that perturbs maternal systems leading to the clinical manifestations of PE. Whilst some of the molecular species driving this stress have been identified, none completely explain the multisystem nature of PE. Syncytiotrophoblast membrane vesicles (STB-EVs) are a potential contributor to this multisystem disorder. STB-EVs are released into the maternal circulation in increasing amounts with advancing gestational age, and this release is further exacerbated with stress. There are good in vitro evidence that STB-EVs are taken up by macrophages and liver cells with additional evidence supporting endothelial cell uptake. STB-EV targeting remains in the early stages of discovery. In this review, we highlight the role of STB-EVs in PE. In relation to current research, we discuss different protocols for ex vivo isolation of STB-EVs, as well as specific issues involving tissue preparation, isolation (some of which may be unique to STB-EVs), and methods for their analysis. We suggest potential solutions for these challenges.
Collapse
|
24
|
Exposure to higher concentrations of exogenous ELABELA causes HTR-8/SVneo trophoblast cell dysfunction: A possible pathogenesis of pre-eclampsia. Pregnancy Hypertens 2022; 30:181-188. [DOI: 10.1016/j.preghy.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/17/2022] [Accepted: 10/09/2022] [Indexed: 11/27/2022]
|
25
|
Calis P, Vojtech L, Hladik F, Gravett MG. A review of ex vivo placental perfusion models: an underutilized but promising method to study maternal-fetal interactions. J Matern Fetal Neonatal Med 2022; 35:8823-8835. [PMID: 34818981 PMCID: PMC9126998 DOI: 10.1080/14767058.2021.2005565] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 02/08/2023]
Abstract
Studying the placenta can provide information about the mechanistic pathways of pregnancy disease. However, analyzing placental tissues and manipulating placental function in real-time during pregnancy is not feasible. The ex vivo placental perfusion model allows observing important aspects of the physiology and pathology of the placenta, while maintaining its viability and functional integrity, and without causing harm to mother or fetus. In this review, we describe and compare setups for this technically complex model and summarize outcomes from various published studies. We hope that our review will encourage wider use of ex vivo placental perfusion, which in turn would generate more knowledge to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Pinar Calis
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Michael G. Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Fetal growth restriction and neonatal-pediatric lung diseases: Vascular mechanistic links and therapeutic directions. Paediatr Respir Rev 2022; 44:19-30. [PMID: 36503648 DOI: 10.1016/j.prrv.2022.09.002] [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: 08/08/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common respiratory sequela of prematurity, and infants born with fetal growth restriction (FGR) are disproportionately represented in BPD statistics, as factors which affect somatic growth may also affect pulmonary growth. Effects of in-utero hypoxia underlying FGR on lung parenchymal architecture predisposing to BPD are well documented, but the pulmonary vascular constructs are not well appreciated. Disruption of angiogenesis during critical periods of lung growth impairs alveolarization, contributing to BPD pathogenesis. Pulmonary artery thickness/stiffness has been noted in FGR in the initial postnatal weeks, and also in well-grown infants with established BPD. The lack of waveform cushioning by the major arteries exposes the pulmonary resistance vessels to higher pulsatile stress, thereby accelerating microvascular disease. Reactive oxygen species, increased sympathetic activity and endothelial dysfunction are common mediators in FGR and BPD; each putative targets for prevention and/or therapeutics using interleukin (IL)-1 receptor antagonist (IL-1Ra), melatonin or inhibition of renin-angiotensin-aldosterone system. While BPD is the archetypal respiratory disease of infancy, effects of FGR on pulmonary function are long-term, extending well into childhood. This narrative links FGR in very/extremely preterm infants with BPD through the vascular affliction as a mechanistic and potentially, therapeutic pathway. Our objectives were to depict the burden of disease for FGR and BPD amongst preterm infants, portray vascular involvement in the placenta in FGR and BPD cohorts, provide high resolution vascular ultrasound information in both cohorts with a view to address therapeutic relevance, and lastly, link this information with paediatric age-group lung diseases.
Collapse
|
27
|
Feng Y, Chen Q, Lau SY, Tsai BW, Groom K, Barrett CJ, Chamley LW. The Blocking of Integrin-Mediated Interactions with Maternal Endothelial Cells Reversed the Endothelial Cell Dysfunction Induced by EVs, Derived from Preeclamptic Placentae. Int J Mol Sci 2022; 23:13115. [PMID: 36361901 PMCID: PMC9657319 DOI: 10.3390/ijms232113115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 08/31/2023] Open
Abstract
Placental extracellular vesicles (EVs) have increasingly been recognized as a major mediator of feto-maternal communication. However, the cellular and molecular mechanisms of the uptake of placental EVs by recipient cells are still not well-understood. We previously reported that placental EVs target a limited number of organs in vivo. In the current study, we investigated the mechanisms underlying the uptake of placental EVs into target cells. Placental EVs were derived from explant cultures of normal or preeclamptic placentae. The mechanisms underlying the uptake of placental EVs were elucidated, using the phagocytosis or endocytosis inhibitor, trypsin-treatment or integrin-blocking peptides. The endothelial cell activation was studied using the monocyte adhesion assay after the preeclamptic EVs exposure, with and/or without treatment with the integrin blocking peptide, YIGSR. The cellular mechanism of the uptake of the placental EVs was time, concentration and energy-dependent and both the phagocytosis and endocytosis were involved in this process. Additionally, proteins on the surface of the placental EVs, including integrins, were involved in the EV uptake process. Furthermore, inhibiting the uptake of preeclamptic EVs with YIGSR, reduced the endothelial cell activation. The interaction between the placental EVs and the recipient cells is mediated by integrins, and the cellular uptake is mediated by a combination of both phagocytosis and endocytosis.
Collapse
Affiliation(s)
- Yourong Feng
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Qi Chen
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Sien Yee Lau
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Bridget W. Tsai
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Katie Groom
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Carolyn J. Barrett
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Lawrence W. Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicles Investigations (HEVI), University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
28
|
Trophoblast Exosomal UCA1 Induces Endothelial Injury through the PFN1-RhoA/ROCK Pathway in Preeclampsia: A Human-Specific Adaptive Pathogenic Mechanism. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2198923. [PMID: 36160709 PMCID: PMC9499815 DOI: 10.1155/2022/2198923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Preeclampsia is regarded as an evolution-related disease that has only been observed in humans and our closest relatives, and the important factor contributing to its pathogenesis is endothelial dysregulation secondary to a stressed placenta. Hypoxia-inducible factor 1 subunit alpha (HIF1α), a highly conserved molecule in virtually all mammals, is regarded as a crucial regulator of the hypoxia adaptation and evolution. Persistent high expression of HIF1α in the placenta is one of the pathogenic mechanisms of preeclampsia. Therefore, human-specific molecules should link increased HIF1α to preeclampsia. We reported that urothelial cancer associated 1 (UCA1) is a potential mediator because it is a human-specific long noncoding RNA (lncRNA) that is upregulated in placental tissues and maternal serum from women with preeclampsia and is regulated by HIF1α. The cellular HIF1α-UCA1 pathway promoted the adaptation of trophoblasts to hypoxia by inducing vascular endothelial growth factor (VEGF) secretion and changes in the levels of key enzymes in glycolysis. On the other hand, circulating exosomal UCA1 secreted from stressed trophoblasts induced vascular endothelial dysfunction, especially excess ROS production, as measured by exosome extraction and a coculture system. At the molecular level, UCA1 physically bound to ubiquitin-specific peptidase 14 (USP14), which is a deubiquitinating enzyme, and UCA1 functioned as a scaffold to recruit USP14 to profilin 1 (PFN1), an actin-binding protein contributing to endothelial abnormalities and vascular diseases. This ternary complex inhibited the ubiquitination-dependent degradation of PFN1 and prolonged its half-life, further activating the RhoA/Rho-kinase (ROCK) pathway to induce ROS production in endothelial cells. Taken together, these observations suggest a role for the evolution-related UCA1 in the HIF1α-induced adaptive pathogenic mechanism of preeclampsia, promoting the survival of hypoxic trophoblasts and injuring maternal endothelial cells.
Collapse
|
29
|
Maligianni I, Yapijakis C, Nousia K, Bacopoulou F, Chrousos G. Exosomes and exosomal non‑coding RNAs throughout human gestation (Review). Exp Ther Med 2022; 24:582. [PMID: 35949320 PMCID: PMC9353550 DOI: 10.3892/etm.2022.11518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/31/2022] [Indexed: 11/06/2022] Open
Abstract
In recent years, research on exosomes and their content has been intensive, which has revealed their important role in cell-to-cell communication, and has implicated exosomal biomolecules in a broad spectrum of physiological processes, as well as in the pathogenesis of various diseases. Pregnancy and its normal progression rely highly on the efficient communication between the mother and the fetus, mainly mediated by the placenta. Recent studies have established the placenta as an important source of circulating exosomes and have demonstrated that exosome release into the maternal circulation gradually increases during pregnancy, starting from six weeks of gestation. This orchestrates maternal-fetal crosstalk, including maternal immune tolerance and pregnancy-associated metabolic adaptations. Furthermore, an increased number of secreted exosomes, along with altered patterns of exosomal non-coding RNAs (ncRNAs), especially microRNAs and long non-coding RNAs (lncRNAs), have been observed in a number of pregnancy complications, such as gestational diabetes mellitus and preeclampsia. The early detection of exosomes and specific exosomal ncRNAs in various biological fluids during pregnancy highlights them as promising candidate biomarkers for the diagnosis, prognosis and treatment of numerous pregnancy disorders in adolescents and adults. The present review aimed to provide insight into the current knowledge regarding the potential, only partially elucidated, role of exosomes and exosomal cargo in the regulation and progression of normal pregnancy, as well as their potential dysregulation and contribution to pathological pregnancy situations.
Collapse
Affiliation(s)
- Ioanna Maligianni
- First Department of Pediatrics, Unit of Orofacial Genetics, ‘Aghia Sophia’ Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Yapijakis
- First Department of Pediatrics, Unit of Orofacial Genetics, ‘Aghia Sophia’ Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantina Nousia
- First Department of Pediatrics, Unit of Orofacial Genetics, ‘Aghia Sophia’ Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
30
|
The Mystery of Exosomes in Gestational Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2169259. [PMID: 35720179 PMCID: PMC9200544 DOI: 10.1155/2022/2169259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 05/31/2022] [Indexed: 11/27/2022]
Abstract
Gestational diabetes mellitus (GDM) is one of the common pregnancy complications, which increases the risk of short-term and long-term adverse consequences in both the mother and offspring. However, the pathophysiological mechanism of GDM is still poorly understood. Inflammation, insulin resistance and oxidative stress are considered critical factors in the occurrence and development of GDM. Although the lifestyle intervention and insulin are the primary treatment, adverse pregnancy outcomes still cannot be ignored. Exosomes have a specific function of carrying biological information, which can transmit information to target cells and play an essential role in intercellular communication. Their possible roles in normal pregnancy and GDM have been widely concerned. The possibility of exosomal cargos as biomarkers of GDM is proposed. This paper reviews the literature in recent years and discusses the role of exosomes in GDM and their possible mechanisms to provide some reference for the prediction, prevention, and treatment of GDM and improve the outcome of pregnancy.
Collapse
|
31
|
Awoyemi T, Iaccarino DA, Motta-Mejia C, Raiss S, Kandzija N, Zhang W, Vatish M. Neuropilin-1 is uniquely expressed on small syncytiotrophoblast extracellular vesicles but not on medium/large vesicles from preeclampsia and normal placentae. Biochem Biophys Res Commun 2022; 619:151-158. [PMID: 35760012 DOI: 10.1016/j.bbrc.2022.06.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Preeclampsia (PE) is a multisystem progressive hypertensive disorder unique to human pregnancy. The placenta is fundamental to its pathogenesis and releases placental factors as well as extracellular vesicles (small and medium/large syncytiotrophoblast extracellular vesicles (STB-EVs)) as a response to syncytiotrophoblast stress such as tissue factor and plasminogen activator inhibitors 1. Neuropilin 1 (NRP-1) is an anti-angiogenic factor involved in development, angiogenesis, arteriogenesis, and vascular permeability. NRP-1 acts as a co-receptor for growth factors such as vascular endothelial growth factor (VEGF), placenta growth factor (PLGF), and epidermal growth factor (EGF). Given the documented pro and anti-angiogenic roles of STB-EVs, we hypothesized that 1) STB-EVs might express NRP-1; and 2) the expression of NRP-1 might differ between normal and preeclampsia STB-EVs. METHODS We isolated STB-EVs (both small and medium/large) from PE and NP placentae using the physiologic ex vivo dual lobe perfusion model. The enriched STB-EVs were characterized by Western blot, transmission electron microscopy (TEM), and nanoparticle tracking analysis (NTA) according to the international society of extracellular vesicles (ISEV) guidelines. We assessed for NRP-1 expression with Western blot (placenta and STB-EVs) and immunohistochemistry (placenta). We performed co-expression analysis for placenta alkaline phosphatase (PLAP - a known STB-EV marker) and NRP-1 with immunoprecipitation followed by Western blot. RESULTS We confirmed NRP-1 expression in NP and PE placenta. We showed that NRP-1 Expression was limited to small syncytiotrophoblast membrane extracellular vesicles (S STB-EVs) but not medium/large STB-EVs and that NRP-1 is co-expressed with PLAP. CONCLUSION Neuropilin-1 is uniquely expressed on small syncytiotrophoblast extracellular vesicles but not on medium/large vesicles from preeclampsia and normal placentae.
Collapse
Affiliation(s)
- Toluwalase Awoyemi
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Daniela A Iaccarino
- Vita-Salute San Raffaele University, Obstetrics and Gynecology Department, Genomic Unit for the Diagnosis of Human Pathologies, Italy
| | - Carolina Motta-Mejia
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Sina Raiss
- S Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Neva Kandzija
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
32
|
Molecular aspects of exposome and metabolic diseases. Mol Aspects Med 2022; 87:101102. [PMID: 35728427 DOI: 10.1016/j.mam.2022.101102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
33
|
Extracellular Vesicles as an Index for Endothelial Injury and Cardiac Dysfunction in a Rodent Model of GDM. Int J Mol Sci 2022; 23:ijms23094970. [PMID: 35563365 PMCID: PMC9101204 DOI: 10.3390/ijms23094970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes mellitus (GDM) increases risk of adverse pregnancy outcomes and maternal cardiovascular complications. It is widely believed that maternal endothelial dysfunction is a critical determinant of these risks, however, connections to maternal cardiac dysfunction and mechanisms of pathogenesis are unclear. Circulating extracellular vesicles (EVs) are emerging biomarkers that may provide insights into the pathogenesis of GDM. We examined the impact of GDM on maternal cardiac and vascular health in a rat model of diet-induced obesity-associated GDM. We observed a >3-fold increase in circulating levels of endothelial EVs (p < 0.01) and von Willebrand factor (p < 0.001) in GDM rats. A significant increase in mitochondrial DNA (mtDNA) within circulating extracellular vesicles was also observed suggesting possible mitochondrial dysfunction in the vasculature. This was supported by nicotinamide adenine dinucleotide deficiency in aortas of GDM mice. GDM was also associated with cardiac remodeling (increased LV mass) and a marked impairment in maternal diastolic function (increased isovolumetric relaxation time [IVRT], p < 0.01). Finally, we observed a strong positive correlation between endothelial EV levels and IVRT (r = 0.57, p < 0.05). In summary, we observed maternal vascular and cardiac dysfunction in rodent GDM accompanied by increased circulating endothelial EVs and EV-associated mitochondrial DNA. Our study highlights a novel method for assessment of vascular injury in GDM and highlights vascular mitochondrial injury as a possible therapeutic target.
Collapse
|
34
|
[Hypertensive disorders of pregnancy in women with COVID-19]. ARCHIVES DES MALADIES DU COEUR ET DES VAISSEAUX. PRATIQUE 2022; 2022:20-24. [PMID: 35153527 PMCID: PMC8818347 DOI: 10.1016/j.amcp.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Redman CW, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the convergence point for multiple pathways. Am J Obstet Gynecol 2022; 226:S907-S927. [PMID: 33546842 DOI: 10.1016/j.ajog.2020.09.047] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Preeclampsia evolves in 2 stages: a placental problem that generates signals to the mother to cause a range of responses that comprise the second stage (preeclampsia syndrome). The first stage of early-onset preeclampsia is poor placentation, which we here call malplacentation. The spiral arteries are incompletely remodeled, leading to later placental malperfusion, relatively early in the second half of pregnancy. The long duration of the first stage (several months) is unsurprisingly associated with fetal growth restriction. The first stage of late-onset preeclampsia, approximately 80% of total cases, is shorter (several weeks) and part of a process that is common to all pregnancies. Placental function declines as it outgrows uterine capacity, with increasing chorionic villous packing, compression of the intervillous space, and fetal hypoxia, and causes late-onset clinical presentations such as "unexplained" stillbirths, late-onset fetal growth restriction, or preeclampsia. The second stages of early- and late-onset preeclampsia share syncytiotrophoblast stress as the most relevant feature that causes the maternal syndrome. Syncytiotrophoblast stress signals in the maternal circulation are probably the most specific biomarkers for preeclampsia. In addition, soluble fms-like tyrosine kinase-1 (mainly produced by syncytiotrophoblast) is the best-known biomarker and is routinely used in clinical practice in many locations. How the stress signals change over time in normal pregnancies indicates that syncytiotrophoblast stress begins on average at 30 to 32 weeks' gestation and progresses to term. At term, syncytiotrophoblast shows increasing markers of stress, including apoptosis, pyroptosis, autophagy, syncytial knots, and necrosis. We label this phenotype the "twilight placenta" and argue that it accounts for the clinical problems of postmature pregnancies. Senescence as a stress response differs in multinuclear syncytiotrophoblast from that of mononuclear cells. Syncytiotrophoblast irreversibly acquires part of the senescence phenotype (cell cycle arrest) when it is formed by cell fusion. The 2 pathways converge on the common pathologic endpoint, syncytiotrophoblast stress, and contribute to preeclampsia subtypes. We highlight that the well-known heterogeneity of the preeclampsia syndrome arises from different pathways to this common endpoint, influenced by maternal genetics, epigenetics, lifestyle, and environmental factors with different fetal and maternal responses to the ensuing insults. This complexity mandates a reassessment of our approach to predicting and preventing preeclampsia, and we summarize research priorities to maximize what we can learn about these important issues.
Collapse
|
36
|
Powell JS, Gandley RE, Lackner E, Dolinish A, Ouyang Y, Powers RW, Morelli AE, Hubel CA, Sadovsky Y. Small extracellular vesicles from plasma of women with preeclampsia increase myogenic tone and decrease endothelium-dependent relaxation of mouse mesenteric arteries. Pregnancy Hypertens 2022; 28:66-73. [DOI: 10.1016/j.preghy.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 10/19/2022]
|
37
|
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
|
38
|
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 I: Health and Normal Physiology: Part I: Health and Normal Physiology. J Extracell Vesicles 2022; 11:e12151. [PMID: 35041249 PMCID: PMC8765331 DOI: 10.1002/jev2.12151] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [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: 12/16/2022] Open
Abstract
Previously thought to be nothing more than cellular debris, extracellular vesicles (EVs) are now known to mediate physiological and pathological functions throughout the body. We now understand more about their capacity to transfer nucleic acids and proteins between distant organs, the interaction of their surface proteins with target cells, and the role of vesicle-bound lipids in health and disease. To date, most observations have been made in reductionist cell culture systems, or as snapshots from patient cohorts. The heterogenous population of vesicles produced in vivo likely act in concert to mediate both beneficial and detrimental effects. EVs play crucial roles in both the pathogenesis of diseases, from cancer to neurodegenerative disease, as well as in the maintenance of system and organ homeostasis. This two-part review draws on the expertise of researchers working in the field of EV biology and aims to cover the functional role of EVs in physiology and pathology. Part I will outline the role of EVs in normal physiology.
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 UniversityHeadington CampusOxfordUK
| | - 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 UniversityHeadington CampusOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - 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 OxfordJohn Radcliffe Hospital, HeadingtonOxfordUK
| |
Collapse
|
39
|
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: 56] [Impact Index Per Article: 28.0] [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
|
40
|
Murugesan S, Hussey H, Saravanakumar L, Sinkey RG, Sturdivant AB, Powell MF, Berkowitz DE. Extracellular Vesicles From Women With Severe Preeclampsia Impair Vascular Endothelial Function. Anesth Analg 2021; 134:713-723. [PMID: 34871190 DOI: 10.1213/ane.0000000000005812] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Preeclampsia (PE) manifesting as hypertension and organ injury is mediated by vascular dysfunction. In biological fluids, extracellular vesicles (EVs) containing microRNA (miRNA), protein, and other cargo released from the placenta may serve as carriers to propagate injury, altering the functional phenotype of endothelial cells. PE has been consistently correlated with increased levels of placenta-derived EVs (pEVs) in maternal circulation. However, whether pEVs impaired endothelial cell function remains to be determined. In this study, we hypothesize that pEVs from pregnant women with severe PE (sPE) impair endothelial function through altered cell signaling. METHODS We obtained plasma samples from women with sPE (n = 14) and normotensive pregnant women (n = 15) for the isolation of EVs. The total number of EV and pEV contribution was determined by quantifying immunoreactive EV-cluster of designation 63 (CD63) and placental alkaline phosphatase (PLAP) as placenta-specific markers, respectively. Vascular endothelial functional assays were determined by cell migration, electric cell-substrate impedance sensing in human aortic endothelial cells (HAECs), and wire myography in isolated blood vessels, preincubated with EVs from normotensive and sPE women. RESULTS Plasma EV and pEV levels were increased in sPE when compared to normotensive without a significant size distribution difference in sPE (108.8 ± 30.2 nm) and normotensive-EVs (101.3 ± 20.3 nm). Impaired endothelial repair and proliferation, reduced endothelial barrier function, reduced endothelial-dependent vasorelaxation, and decreased nitrite level indicate that sPE-EVs induced vascular endothelial dysfunction. Moreover, sPE-EVs significantly downregulated endothelial nitric oxide synthase (eNOS and p-eNOS) when compared to normotensive-EV. CONCLUSIONS EVs from sPE women impair endothelial-dependent vascular functions in vitro.
Collapse
Affiliation(s)
- Saravanakumar Murugesan
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| | - Hanna Hussey
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| | - Lakshmi Saravanakumar
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Adam B Sturdivant
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| | - Mark F Powell
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| | - Dan E Berkowitz
- From the Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine
| |
Collapse
|
41
|
Chuaiphichai S, Yu GZ, Tan CM, Whiteman C, Douglas G, Dickinson Y, Drydale EN, Appari M, Zhang W, Crabtree MJ, McNeill E, Hale AB, Lewandowski AJ, Alp NJ, Vatish M, Leeson P, Channon KM. Endothelial GTPCH (GTP Cyclohydrolase 1) and Tetrahydrobiopterin Regulate Gestational Blood Pressure, Uteroplacental Remodeling, and Fetal Growth. Hypertension 2021; 78:1871-1884. [PMID: 34689592 PMCID: PMC8577301 DOI: 10.1161/hypertensionaha.120.17646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Grace Z. Yu
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine (G.Z.Y., C.M.J.T., A.J.L., P.L.), University of Oxford, United Kingdom
| | - Cheryl M.J. Tan
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine (G.Z.Y., C.M.J.T., A.J.L., P.L.), University of Oxford, United Kingdom
| | - Christopher Whiteman
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine (G.Z.Y., C.M.J.T., A.J.L., P.L.), University of Oxford, United Kingdom
- Nuffield Department of Women’s and Reproductive Health (W.Z., M.V.), University of Oxford, United Kingdom
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service Foundation Trust, John Radcliffe Hospital, United Kingdom (M.V., K.M.C.)
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Yasmin Dickinson
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Edward N. Drydale
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Mahesh Appari
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Wei Zhang
- Nuffield Department of Women’s and Reproductive Health (W.Z., M.V.), University of Oxford, United Kingdom
| | - Mark J. Crabtree
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Eileen McNeill
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Ashley B. Hale
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Adam J. Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine (G.Z.Y., C.M.J.T., A.J.L., P.L.), University of Oxford, United Kingdom
| | - Nicholas J. Alp
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
| | - Manu Vatish
- Nuffield Department of Women’s and Reproductive Health (W.Z., M.V.), University of Oxford, United Kingdom
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service Foundation Trust, John Radcliffe Hospital, United Kingdom (M.V., K.M.C.)
| | - Paul Leeson
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine (G.Z.Y., C.M.J.T., A.J.L., P.L.), University of Oxford, United Kingdom
| | - Keith M. Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.C., C.W., G.D., Y.D., E.N.D., M.A., M.J.C., E.M., A.B.H., N.J.A., K.M.C.)
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service Foundation Trust, John Radcliffe Hospital, United Kingdom (M.V., K.M.C.)
| |
Collapse
|
42
|
Kohli S, Isermann B. Crosstalk between inflammation and coagulation: Focus on pregnancy related complications. THROMBOSIS UPDATE 2021. [DOI: 10.1016/j.tru.2021.100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
43
|
Erlandsson L, Masoumi Z, Hansson LR, Hansson SR. The roles of free iron, heme, haemoglobin, and the scavenger proteins haemopexin and alpha-1-microglobulin in preeclampsia and fetal growth restriction. J Intern Med 2021; 290:952-968. [PMID: 34146434 DOI: 10.1111/joim.13349] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a complex pregnancy syndrome characterised by maternal hypertension and organ damage after 20 weeks of gestation and is associated with an increased risk of cardiovascular disease later in life. Extracellular haemoglobin (Hb) and its metabolites heme and iron are highly toxic molecules and several defence mechanisms have evolved to protect the tissue. OBJECTIVES We will discuss the roles of free iron, heme, Hb, and the scavenger proteins haemopexin and alpha-1-microglobulin in pregnancies complicated by PE and fetal growth restriction (FGR). CONCLUSION In PE, oxidative stress causes syncytiotrophoblast (STB) stress and increased shedding of placental STB-derived extracellular vesicles (STBEV). The level in maternal circulation correlates with the severity of hypertension and supports the involvement of STBEVs in causing maternal symptoms in PE. In PE and FGR, iron homeostasis is changed, and iron levels significantly correlate with the severity of the disease. The normal increase in plasma volume taking place during pregnancy is less for PE and FGR and therefore have a different impact on, for example, iron concentration, compared to normal pregnancy. Excess iron promotes ferroptosis is suggested to play a role in trophoblast stress and lipotoxicity. Non-erythroid α-globin regulates vasodilation through the endothelial nitric oxide synthase pathway, and hypoxia-induced α-globin expression in STBs in PE placentas is suggested to contribute to hypertension in PE. Underlying placental pathology in PE with and without FGR might be amplified by iron and heme overload causing oxidative stress and ferroptosis. As the placenta becomes stressed, the release of STBEVs increases and affects the maternal vasculature.
Collapse
Affiliation(s)
- Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zahra Masoumi
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lucas R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Obstetrics and Gynecology, Skåne University Hospital, Lund/Malmö, Sweden
| |
Collapse
|
44
|
Palei AC, Granger JP, Spradley FT. Placental Ischemia Says "NO" to Proper NOS-Mediated Control of Vascular Tone and Blood Pressure in Preeclampsia. Int J Mol Sci 2021; 22:ijms222011261. [PMID: 34681920 PMCID: PMC8541176 DOI: 10.3390/ijms222011261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022] Open
Abstract
In this review, we first provide a brief overview of the nitric oxide synthase (NOS) isoforms and biochemistry. This is followed by describing what is known about NOS-mediated blood pressure control during normal pregnancy. Circulating nitric oxide (NO) bioavailability has been assessed by measuring its metabolites, nitrite (NO2) and/or nitrate (NO3), and shown to rise throughout normal pregnancy in humans and rats and decline postpartum. In contrast, placental malperfusion/ischemia leads to systemic reductions in NO bioavailability leading to maternal endothelial and vascular dysfunction with subsequent development of hypertension in PE. We end this article by describing emergent risk factors for placental malperfusion and ischemic disease and discussing strategies to target the NOS system therapeutically to increase NO bioavailability in preeclamptic patients. Throughout this discussion, we highlight the critical importance that experimental animal studies have played in our current understanding of NOS biology in normal pregnancy and their use in finding novel ways to preserve this signaling pathway to prevent the development, treat symptoms, or reduce the severity of PE.
Collapse
Affiliation(s)
- Ana C. Palei
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Joey P. Granger
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Frank T. Spradley
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS 39216, USA;
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA;
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
45
|
León J, Acurio J, Bergman L, López J, Karin Wikström A, Torres-Vergara P, Troncoso F, Castro FO, Vatish M, Escudero C. Disruption of the Blood-Brain Barrier by Extracellular Vesicles From Preeclampsia Plasma and Hypoxic Placentae: Attenuation by Magnesium Sulfate. Hypertension 2021; 78:1423-1433. [PMID: 34601964 DOI: 10.1161/hypertensionaha.121.17744] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- José León
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile (J. Leon,.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
- Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
- Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.)
| | - Juán López
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
| | - Anna Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Chile (P.T.-V.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepcion, Chillan, Chile (F.O.C.)
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom (M.V.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| |
Collapse
|
46
|
Munjas J, Sopić M, Stefanović A, Košir R, Ninić A, Joksić I, Antonić T, Spasojević-Kalimanovska V, Prosenc Zmrzljak U. Non-Coding RNAs in Preeclampsia-Molecular Mechanisms and Diagnostic Potential. Int J Mol Sci 2021; 22:10652. [PMID: 34638993 PMCID: PMC8508896 DOI: 10.3390/ijms221910652] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia (PE) is a leading cause of maternal and neonatal morbidity and mortality worldwide. Defects in trophoblast invasion, differentiation of extravillous trophoblasts and spiral artery remodeling are key factors in PE development. Currently there are no predictive biomarkers clinically available for PE. Recent technological advancements empowered transcriptome exploration and led to the discovery of numerous non-coding RNA species of which microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are the most investigated. They are implicated in the regulation of numerous cellular functions, and as such are being extensively explored as potential biomarkers for various diseases. Altered expression of numerous lncRNAs and miRNAs in placenta has been related to pathophysiological processes that occur in preeclampsia. In the following text we offer summary of the latest knowledge of the molecular mechanism by which lnRNAs and miRNAs (focusing on the chromosome 19 miRNA cluster (C19MC)) contribute to pathophysiology of PE development and their potential utility as biomarkers of PE, with special focus on sample selection and techniques for the quantification of lncRNAs and miRNAs in maternal circulation.
Collapse
Affiliation(s)
- Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Aleksandra Stefanović
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Rok Košir
- BIA Separations CRO, Labena Ltd., Street Verovškova 64, 1000 Ljubljana, Slovenia;
| | - Ana Ninić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Ivana Joksić
- Genetic Laboratory Department, Obstetrics and Gynaecology Clinic “Narodni Front”, Street Kraljice Natalije 62, 11000 Belgrade, Serbia;
| | - Tamara Antonić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Vesna Spasojević-Kalimanovska
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | | |
Collapse
|
47
|
Procoagulant Extracellular Vesicles Alter Trophoblast Differentiation in Mice by a Thrombo-Inflammatory Mechanism. Int J Mol Sci 2021; 22:ijms22189873. [PMID: 34576036 PMCID: PMC8466022 DOI: 10.3390/ijms22189873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Procoagulant extracellular vesicles (EV) and platelet activation have been associated with gestational vascular complications. EV-induced platelet-mediated placental inflammasome activation has been shown to cause preeclampsia-like symptoms in mice. However, the effect of EV-mediated placental thrombo-inflammation on trophoblast differentiation remains unknown. Here, we identify that the EV-induced thrombo-inflammatory pathway modulates trophoblast morphology and differentiation. EVs and platelets reduce syncytiotrophoblast differentiation while increasing giant trophoblast and spongiotrophoblast including the glycogen-rich cells. These effects are platelet-dependent and mediated by the NLRP3 inflammasome. In humans, inflammasome activation was negatively correlated with trophoblast differentiation marker GCM1 and positively correlated with blood pressure. These data identify a crucial role of EV-induced placental thrombo-inflammation on altering trophoblast differentiation and suggest platelet activation or inflammasome activation as a therapeutic target in order to achieve successful placentation.
Collapse
|
48
|
Rajaratnam N, Ditlevsen NE, Sloth JK, Bæk R, Jørgensen MM, Christiansen OB. Extracellular Vesicles: An Important Biomarker in Recurrent Pregnancy Loss? J Clin Med 2021; 10:jcm10122549. [PMID: 34207656 PMCID: PMC8226857 DOI: 10.3390/jcm10122549] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Recurrent pregnancy loss (RPL) has an estimated incidence of 1–3% of all couples. The etiology is considered to be multifactorial. Extracellular vesicles (EVs) take part in numerous different physiological processes and their contents show the originating cell and pathophysiological states in different diseases. In pregnancy disorders, changes can be seen in the composition, bioactivity and concentration of placental and non-placental EVs. RPL patients have an increased risk of pregnancy complications. The aim of this prospective study was to examine whether measuring different specific EV markers in plasma before and during pregnancy could be used as predictors of pregnancy loss (PL) in women with RPL. Thirty-one RPL patients were included in this study; 25 had a live birth (LB group) and six had a new PL (PL group). Five blood samples were obtained, one before achieved pregnancy and the others in gestational week 6, 8, 10 and 16. Moreover, some of the patients received intravenous immunoglobulin (IVIG) infusions as part of treatment, and it was also examined whether this treatment influenced the EV levels. Seventeen EV markers specific for the immune system, coagulation, placenta and hypoxia were analyzed in the samples with EV Array, a method able to capture small EVs by using an antibody panel targeting membrane proteins. Comparing the LB and PL groups, one EV marker, CD9, showed a significant increase from before pregnancy to gestational week 6 in the PL group. The changes in the other 16 markers were nonsignificant. One case of late-onset PL showed steeply increasing levels, with sudden decrease after gestational week 10 in nine of 17 markers. Moreover, there was an overall increase of all 17 markers after IVIG treatment in the LB group, which was significant in 15 of the markers. Whether increases in EVs positive for CD9 characterize RPL patients who subsequently miscarry should be investigated in future larger studies.
Collapse
Affiliation(s)
- Nina Rajaratnam
- Department of Obstetrics and Gynecology, Aalborg University Hospital, 9000 Aalborg, Denmark; (N.E.D.); (O.B.C.)
- Correspondence:
| | - Nadja E. Ditlevsen
- Department of Obstetrics and Gynecology, Aalborg University Hospital, 9000 Aalborg, Denmark; (N.E.D.); (O.B.C.)
| | - Jenni K. Sloth
- Department of Clinical Immunology, Aalborg University Hospital, 9000 Aalborg, Denmark; (J.K.S.); (R.B.); (M.M.J.)
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, 9000 Aalborg, Denmark; (J.K.S.); (R.B.); (M.M.J.)
| | - Malene M. Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, 9000 Aalborg, Denmark; (J.K.S.); (R.B.); (M.M.J.)
- Department of Clincial Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Ole B. Christiansen
- Department of Obstetrics and Gynecology, Aalborg University Hospital, 9000 Aalborg, Denmark; (N.E.D.); (O.B.C.)
- Clinical Institute, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
49
|
Martins-Marques T, Rodriguez-Sinovas A, Girao H. Cellular crosstalk in cardioprotection: Where and when do reactive oxygen species play a role? Free Radic Biol Med 2021; 169:397-409. [PMID: 33892116 DOI: 10.1016/j.freeradbiomed.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
A well-balanced intercellular communication between the different cells within the heart is vital for the maintenance of cardiac homeostasis and function. Despite remarkable advances on disease management and treatment, acute myocardial infarction remains the major cause of morbidity and mortality worldwide. Gold standard reperfusion strategies, namely primary percutaneous coronary intervention, are crucial to preserve heart function. However, reestablishment of blood flow and oxygen levels to the infarcted area are also associated with an accumulation of reactive oxygen species (ROS), leading to oxidative damage and cardiomyocyte death, a phenomenon termed myocardial reperfusion injury. In addition, ROS signaling has been demonstrated to regulate multiple biological pathways, including cell differentiation and intercellular communication. Given the importance of cell-cell crosstalk in the coordinated response after cell injury, in this review, we will discuss the impact of ROS in the different forms of inter- and intracellular communication, as well as the role of gap junctions, tunneling nanotubes and extracellular vesicles in the propagation of oxidative damage in cardiac diseases, particularly in the context of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Antonio Rodriguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall D'Hebron Institut de Recerca (VHIR), Vall D'Hebron Hospital Universitari, Vall D'Hebron Barcelona Hospital Campus, Passeig Vall D'Hebron, 119-129, 08035, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Henrique Girao
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
50
|
Mahmood S, Younas H, Younus A, Nathenial S. A narrative review on the role of folate-mediated one-carbon metabolism and its associated gene polymorphisms in posing risk to preeclampsia. Clin Exp Hypertens 2021; 43:487-504. [PMID: 34053381 DOI: 10.1080/10641963.2021.1916942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Preeclampsia (PE) presents a major obstetrical problem for mother and fetus which is characterized by the onset of hypertension and proteinuria in formerly normotensive women. Altered folate-mediated one-carbon metabolism is one of the factors for PE development either due to nutritional insufficiencies such as folate deficiency or polymorphisms in genes that code for the key enzymes of the cycle. Commonly, there are four genes in the cycle whose polymorphisms have been described in relation to PE. These factors could cause elevation of homocysteine; the toxic metabolite, which subsequently leads to the development of PE. Sufficient levels of folate have been considered important during pregnancy and may reduce the risk of development of PE. This review aims at discussing genetic polymorphisms and nutritional deficiencies as probable predisposing factors and suggests considering fetal genotypes, varied ethnicities, and interaction of various other factors involved to render better conclusiveness to the present studies.
Collapse
Affiliation(s)
- Sadia Mahmood
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Hooria Younas
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Amna Younus
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sammar Nathenial
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| |
Collapse
|