1
|
Wang T, Huang W, Gao X, Deng Y, Huang J. Single extracellular vesicle research: From cell population to a single cell. Biochem Biophys Res Commun 2024; 734:150439. [PMID: 39083971 DOI: 10.1016/j.bbrc.2024.150439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Extracellular vesicles (EVs) are secreted by cells with a membrane structure and complex components such as DNA, RNA and proteins. These biomolecules play an important role in cell communication, cell proliferation, cell migration, vascularization, immune response and other physiological and pathological processes. Most current research on EVs focused on populations of EVs. Heterogeneity of EVs is neglected. Considering the heterogeneity of single EVs may offer critical molecular insights into cell-cell interactions, it is necessary to enhance our understanding about molecular characteristics from EVs derived from cell population to a single EV of derived from a single cell. This transformation is expected to provide a new insight into the understanding of cellular biology and the accurate description of the law of disease progress. In this article, we review the current research progress of single EV analysis technology for single EVs derived from cell population (SECP) and discuss its main applications in biological and clinical medicine research. After that, we propose the development direction, main difficulties and application prospect of single EV analysis technology for single EVs derived from single cells (SESC) according to our own research work, to provide new perspectives for the field of EV research.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xu Gao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Musa E, Salazar-Petres E, Vatish M, Levitt N, Sferruzzi-Perri AN, Matjila MJ. Kisspeptin signalling and its correlation with placental ultrastructure and clinical outcomes in pregnant South African women with obesity and gestational diabetes. Placenta 2024; 154:49-59. [PMID: 38878622 DOI: 10.1016/j.placenta.2024.05.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 09/03/2024]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is a major pregnancy metabolic disorder and is strongly linked with obesity. Kisspeptin is a hormone that increases several thousand-fold in the maternal circulation during human pregnancy, with placenta as its main source. Studies have suggested that kisspeptin regulates trophoblast invasion and promotes pancreatic insulin secretion and peripheral insulin sensitivity. METHODS In a well-characterized cohort of pregnant South African women and molecular and histological techniques, this study explored the impact and interaction of maternal obesity and GDM on kisspeptin (KISS1) signalling in relation to placental morphology and maternal and neonatal parameters. RESULTS We found that GDM had no effect on placental KISS1 and KISS1R (KISS1 receptor) mRNA and/or protein expression. However, obesity reduced placental KISS1R mRNA expression even though overall KISS1 protein abundance or localization was not different from the non-obese group. Maternal and cord circulating KISS1 concentrations did not vary with obesity or GDM, but maternal circulating KISS1 was positively correlated with placenta weight in non-GDM obese women, and negatively correlated with placental intervillous space volume in non-GDM non-obese women. Cord serum KISS1 was positively correlated with infant weight in GDM obese women, but negatively correlated with maternal BMI in the non-obese GDM group. Placental syncytiotrophoblast extracellular vesicles exhibited detectable KISS1 and its abundance was ∼50 % lower in those from obese GDM compared to non-GDM women. DISCUSSION This study shows maternal obesity and GDM can modulate placental kisspeptin signalling and placental morphological development with potential pathophysiological implications for clinically-relevant pregnancy and perinatal outcomes.
Collapse
Affiliation(s)
- Ezekiel Musa
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa; Department of Internal Medicine, Kaduna State University, Kaduna, Nigeria
| | - Esteban Salazar-Petres
- Carrera de Obstetricia, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Valdivia, Chile
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Naomi Levitt
- Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Mushi J Matjila
- Department of Obstetrics and Gynaecology, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
3
|
Sokolov D, Gorshkova A, Tyshchuk E, Grebenkina P, Zementova M, Kogan I, Totolian A. Large Extracellular Vesicles Derived from Natural Killer Cells Affect the Functions of Monocytes. Int J Mol Sci 2024; 25:9478. [PMID: 39273424 PMCID: PMC11395174 DOI: 10.3390/ijms25179478] [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: 06/03/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Communication between natural killer cells (NK cells) and monocytes/macrophages may play an important role in immunomodulation and regulation of inflammatory processes. The aim of this research was to investigate the impact of NK cell-derived large extracellular vesicles on monocyte function because this field is understudied. We studied how NK-cell derived large extracellular vesicles impact on THP-1 cells characteristics after coculturing: phenotype, functions were observed with flow cytometry. In this study, we demonstrated the ability of large extracellular vesicles produced by NK cells to integrate into the membranes of THP-1 cells and influence the viability, phenotype, and functional characteristics of the cells. The results obtained demonstrate the ability of large extracellular vesicles to act as an additional component in the immunomodulatory activity of NK cells in relation to monocytes.
Collapse
Affiliation(s)
- Dmitry Sokolov
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
- Saint-Petersburg Pasteur Institute, 197101 St. Petersburg, Russia
| | - Alina Gorshkova
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
| | - Elizaveta Tyshchuk
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
- Saint-Petersburg Pasteur Institute, 197101 St. Petersburg, Russia
| | - Polina Grebenkina
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
- Saint-Petersburg Pasteur Institute, 197101 St. Petersburg, Russia
| | - Maria Zementova
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
- Saint-Petersburg Pasteur Institute, 197101 St. Petersburg, Russia
| | - Igor Kogan
- Federal State Budgetary Scientific Institution "The Research Institute of Obstetrics, Gynecology and Reproductology Named after D.O. Ott", 199034 St. Petersburg, Russia
| | - Areg Totolian
- Saint-Petersburg Pasteur Institute, 197101 St. Petersburg, Russia
| |
Collapse
|
4
|
Fakouri A, Razavi ZS, Mohammed AT, Hussein AHA, Afkhami H, Hooshiar MH. Applications of mesenchymal stem cell-exosome components in wound infection healing: new insights. BURNS & TRAUMA 2024; 12:tkae021. [PMID: 39139205 PMCID: PMC11319788 DOI: 10.1093/burnst/tkae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/12/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024]
Abstract
The healing process at a wound is made up of many types of cells, growth factors, the extracellular matrix, nerves and blood vessels all interacting with each other in complex and changing ways. Microbial colonization and proliferation are possible at the place of injury, which makes infection more likely. Because of this, any cut has a chance of getting an infection. Researchers have found that wound infections make patients more upset and cost the healthcare system a lot of money. Surgical site infections happen a lot to people who have recently had surgery. This study shows that such surgical infection is linked to a high rate of illness and death. This is shown by the fact that 25% of patients get serious sepsis and need to be transferred to an intensive care unit. In both animal models and people, mesenchymal stem cells (MSCs) play an active role in all stages of wound healing and have positive effects. Exosomes are one of the main things MSCs release. They have effects that are similar to those of the parent MSCs. Various effector proteins, messenger RNA and microRNAs can be transported by extracellular vesicles to control the activity of target cells. This has a big impact on the healing process. These results suggest that using MSC-exosomes as a new type of cell-free therapy could be a better and safer option than whole cell therapy. This review is mostly about how to use parts of MSC-exosomes to help wound infections heal.
Collapse
Affiliation(s)
- Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad 6813833946, Iran
| | - Zahra-Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | |
Collapse
|
5
|
Sokolov I. Ultrabright fluorescent particles via physical encapsulation of fluorescent dyes in mesoporous silica: a mini-review. NANOSCALE 2024; 16:10994-11004. [PMID: 38771589 DOI: 10.1039/d4nr00800f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Harnessing the power of mesoporous silica to encapsulate organic fluorescent dyes has led to the creation of an extraordinary class of nanocomposite photonic materials. These materials stand out for their ability to produce the brightest fluorescent particles known today, surpassing even the luminosity of quantum dots of similar spectrum and size. The synthesis of these materials offers precise control over the shape and size of the particles, ranging from the nano to the multi-micron scale. Just physical encapsulation of the dyes opens new possibilities for mixing different dyes within individual particles, paving the way for nearly limitless multiplexing capabilities. Moreover, this approach lays the groundwork for the development of highly sensitive sensors capable of detecting subtle changes in temperature and acidity at the nanoscale, among other parameters. This mini-review highlights the mechanism of synthesis, explains the nature of ultrabrightness, and describes the recent advancements and future prospects in the field of ultrabright fluorescent mesoporous silica particles, showcasing their potential for various applications.
Collapse
Affiliation(s)
- Igor Sokolov
- Department of Mechanical Engineering, Tufts University, Medford, Massachusetts, USA.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
- Department of Physics, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
6
|
Nair S, Razo-Azamar M, Jayabalan N, Dalgaard LT, Palacios-González B, Sørensen A, Kampmann U, Handberg A, Carrion F, Salomon C. Advances in extracellular vesicles as mediators of cell-to-cell communication in pregnancy. Cytokine Growth Factor Rev 2024; 76:86-98. [PMID: 38233286 DOI: 10.1016/j.cytogfr.2023.12.004] [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: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Cell-to-cell communication mediated by Extracellular Vesicles (EVs) is a novel and emerging area of research, especially during pregnancy, in which placenta derived EVs can facilitate the feto-maternal communication. EVs comprise a heterogeneous group of vesicle sub-populations with diverse physical and biochemical characteristics and originate by specific biogenesis mechanisms. EVs transfer molecular cargo (including proteins, nucleic acids, and lipids) between cells and are critical mediators of cell communication. There is growing interest among researchers to explore into the molecular cargo of EVs and their functions in a physiological and pathological context. For example, inflammatory mediators such as cytokines are shown to be released in EVs and EVs derived from immune cells play key roles in mediating the immune response as well as immunoregulatory pathways. Pregnancy complications such as gestational diabetes mellitus, preeclampsia, intrauterine growth restriction and preterm birth are associated with altered levels of circulating EVs, with differential EV cargo and bioactivity in target cells. This implicates the intriguing roles of EVs in reprogramming the maternal physiology during pregnancy. Moreover, the capacity of EVs to carry bioactive molecules makes them a promising tool for biomarker development and targeted therapies in pregnancy complications. This review summarizes the physiological and pathological roles played by EVs in pregnancy and pregnancy-related disorders and describes the potential of EVs to be translated into clinical applications in the diagnosis and treatment of pregnancy complications.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia.
| | - Melissa Razo-Azamar
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; Laboratorio de Envejecimiento Saludable del Instituto Nacional de Medicina Genómica (INMEGEN) en el Centro de Investigación sobre Envejecimiento (CIE-CINVESTAV Sede Sur), CDMX, 14330, Mexico
| | - Nanthini Jayabalan
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
| | | | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable del Instituto Nacional de Medicina Genómica (INMEGEN) en el Centro de Investigación sobre Envejecimiento (CIE-CINVESTAV Sede Sur), CDMX, 14330, Mexico
| | - Anne Sørensen
- Department of Obstetrics and Gynecology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, and Department of Clinical Medicine, Aarhus University, Denmark
| | - Aase Handberg
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
7
|
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
|
8
|
Leung J, Pollalis D, Nair GKG, Bailey JK, Pennington BO, Khan AI, Kelly KR, Yeh AK, Sundaram KS, Clegg DO, Peng CC, Xu L, Lee SY. Isolation and Characterization of Extracellular Vesicles Through Orthogonal Approaches for the Development of Intraocular EV Therapy. Invest Ophthalmol Vis Sci 2024; 65:6. [PMID: 38466285 PMCID: PMC10929743 DOI: 10.1167/iovs.65.3.6] [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: 12/01/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose Isolating extracellular vesicles (EVs) with high yield, replicable purity, and characterization remains a bottleneck in the development of EV therapeutics. To address these challenges, the current study aims to establish the necessary framework for preclinical and clinical studies in the development of stem cell-derived intraocular EV therapeutics. Methods Small EVs (sEVs) were separated from the conditioned cell culture medium (CCM) of the human embryogenic stem cell-derived fully polarized retinal pigment epithelium (hESC-RPE-sEV) by a commercially available microfluidic tangential flow filtration (TFF) device ExoDisc (ED) or differential ultracentrifugation (dUC). The scaling and concentration capabilities and purity of recovered sEVs were assessed. Size, number, and surface markers of sEVs were determined by orthogonal approaches using multiple devices. Results ED yielded higher numbers of sEVs, ranging from three to eight times higher depending on the measurement device, compared to dUC using the same 5 mL of CCM input. Within the same setting, the purity of ED-recovered hESC-RPE-sEVs was higher than that for dUC-recovered sEVs. ED yielded a higher concentration of particles, which is strongly correlated with the input volume, up to 10 mL (r = 0.98, P = 0.016). Meanwhile, comprehensive characterization profiles of EV surface markers between ED- and dUC-recovered hESC-RPE-sEVs were compatible. Conclusions Our study supports TFF as a valuable strategy for separating sEVs for the development of intraocular EV therapeutics. However, there is a growing need for diverse devices to optimize TFF for use in EV preparation. Using orthogonal approaches in EV characterization remains ideal for reliably characterizing heterogeneous EV.
Collapse
Affiliation(s)
- Justin Leung
- USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
| | - Dimitrios Pollalis
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Gopa K. G. Nair
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Jeffrey K. Bailey
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Britney O. Pennington
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Amir I. Khan
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kaitlin R. Kelly
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Ashley K. Yeh
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- College of Creative Studies, Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kartik S. Sundaram
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Dennis O. Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Chen-Ching Peng
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Liya Xu
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Sun Young Lee
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
9
|
Kandzija N, Payne S, Cooke WR, Seedat F, Fischer R, Vatish M. Protein Profiling of Placental Extracellular Vesicles in Gestational Diabetes Mellitus. Int J Mol Sci 2024; 25:1947. [PMID: 38396626 PMCID: PMC10887986 DOI: 10.3390/ijms25041947] [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: 01/01/2024] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Throughout pregnancy, some degree of insulin resistance is necessary to divert glucose towards the developing foetus. In gestational diabetes mellitus (GDM), insulin resistance is exacerbated in combination with insulin deficiency, causing new-onset maternal hyperglycaemia. The rapid reversal of insulin resistance following delivery strongly implicates the placenta in GDM pathogenesis. In this case-control study, we investigated the proteomic cargo of human syncytiotrophoblast-derived extracellular vesicles (STBEVs), which facilitate maternal-fetal signalling during pregnancy, in a UK-based cohort comprising patients with a gestational age of 38-40 weeks. Medium/large (m/l) and small (s) STBEVs were isolated from GDM (n = 4) and normal (n = 5) placentae using ex vivo dual-lobe perfusion and subjected to mass spectrometry. Bioinformatics were used to identify differentially carried proteins and mechanistic pathways. In m/lSTBEVs, 56 proteins were differently expressed while in sSTBEVs, no proteins reached statistical difference. Differences were also observed in the proteomic cargo between m/lSTBEVs and sSTBEVs, indicating that the two subtypes of STBEVs may have divergent modes of action and downstream effects. In silico functional enrichment analysis of differentially expressed proteins in m/lSTBEVs from GDM and normal pregnancy found positive regulation of cytoskeleton organisation as the most significantly enriched biological process. This work presents the first comparison of two populations of STBEVs' protein cargos (m/l and sSTBEVs) from GDM and normal pregnancy isolated using placenta perfusion. Further investigation of differentially expressed proteins may contribute to an understanding of GDM pathogenesis and the development of novel diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Neva Kandzija
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; (N.K.); (S.P.); (W.R.C.); (F.S.)
| | - Sophie Payne
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; (N.K.); (S.P.); (W.R.C.); (F.S.)
| | - William R. Cooke
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; (N.K.); (S.P.); (W.R.C.); (F.S.)
| | - Faheem Seedat
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; (N.K.); (S.P.); (W.R.C.); (F.S.)
| | - Roman Fischer
- Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, UK;
| | - Manu Vatish
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; (N.K.); (S.P.); (W.R.C.); (F.S.)
| |
Collapse
|
10
|
Barnes MVC, Pantazi P, Holder B. Circulating extracellular vesicles in healthy and pathological pregnancies: A scoping review of methodology, rigour and results. J Extracell Vesicles 2023; 12:e12377. [PMID: 37974377 PMCID: PMC10654380 DOI: 10.1002/jev2.12377] [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: 05/05/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in pregnancy, revealed by the presence of placental-derived EVs in maternal blood, their in vitro functionality, and their altered cargo in pregnancy pathologies. These EVs are thought to be involved in the development of pregnancy pathologies, such as pre-eclampsia, pre-term birth, and fetal growth restriction, and have been suggested as a source of biomarkers for gestational diseases. However, to accurately interpret their function and biomarker potential, it is necessary to critically evaluate the EV isolation and characterization methodologies used in pregnant cohorts. In this systematic scoping review, we collated the results from 152 studies that have investigated EVs in the blood of pregnant women, and provide a detailed analysis of the EV isolation and characterization methodologies used. Our findings indicate an overall increase in EV concentrations in pregnant compared to non-pregnant individuals, an increased EV count as gestation progresses, and an increased EV count in some pregnancy pathologies. We highlight the need for improved standardization of methodology, greater focus on gestational changes in EV concentrations, and further investigations into the functionality of EVs. Our review suggests that EVs hold great promise as diagnostic and translational tools for gestational diseases. However, to fully realize their potential, it is crucial to improve the standardization and reliability of EV isolation and characterization methodologies, and to gain a better understanding of their functional roles in pregnancy pathologies.
Collapse
Affiliation(s)
- Megan V. C. Barnes
- Institute of Reproductive and Developmental Biology, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
| | - Paschalia Pantazi
- Institute of Reproductive and Developmental Biology, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
| | - Beth Holder
- Institute of Reproductive and Developmental Biology, Department of MetabolismDigestion and Reproduction, Imperial College LondonLondonUK
| |
Collapse
|
11
|
Erlandsson L, Ohlsson L, Masoumi Z, Rehnström M, Cronqvist T, Edvinsson L, Hansson SR. Preliminary evidence that blocking the uptake of placenta-derived preeclamptic extracellular vesicles protects the vascular endothelium and prevents vasoconstriction. Sci Rep 2023; 13:18425. [PMID: 37891193 PMCID: PMC10611745 DOI: 10.1038/s41598-023-45830-9] [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: 07/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023] Open
Abstract
Preeclampsia (PE) is a pregnancy syndrome characterized by hypertension and organ damage manifesting after 20 gestational weeks. The etiology is of multifactorial origin, where placental stress causes increased levels of placenta-derived extracellular vesicles (STBEVs) in the maternal circulation, shown to cause inflammation, endothelial activation, vasoconstriction, and anti-angiogenic activity. General endothelial dysfunction is believed to be initiated by endothelial insult during pregnancy that alters vascular function resulting in increased arterial stiffness, cardiac dysfunction, and increased risk of cardiovascular disease later in life. We compared the effect of normal and PE derived STBEVs in vitro on vascular contractility of human subcutaneous arteries using wire myography. Cellular structures of exposed vessels were investigated by transmission electron microscopy. We explored strategies to pharmacologically block the effects of the STBEVs on human vessels. The PE STBEVs caused significantly stronger angiotensin II-mediated contractions and extended structural damage to human subcutaneous arteries compared to normal STBEVs. These negative effects could be reduced by blocking vesicle uptake by endothelial cells, using chlorpromazine or specific antibodies towards the LOX-1 receptor. The therapeutic potential of blocking vesicle uptake should be further explored, to reduce the permanent damage caused on the vasculature during PE pregnancy to prevent future cardiovascular risk.
Collapse
Affiliation(s)
- Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden.
| | - Lena Ohlsson
- Experimental Vascular Research, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zahra Masoumi
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Mimmi Rehnström
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Tina Cronqvist
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Lars Edvinsson
- Experimental Vascular Research, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Skåne University Hospital, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
- Skåne University Hospital, Lund, Sweden
| |
Collapse
|
12
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke CLM, Davidge ST. Placenta-Derived Extracellular Vesicles From Preeclamptic Pregnancies Impair Vascular Endothelial Function via Lectin-Like Oxidized LDL Receptor-1. Hypertension 2023; 80:2226-2238. [PMID: 37615097 DOI: 10.1161/hypertensionaha.123.21205] [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: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Preeclampsia is a complex syndrome that includes maternal vascular dysfunction. Syncytiotrophoblast-derived extracellular vesicles from preeclampsia placentas (preeclampsia-STBEVs) were shown to induce endothelial dysfunction, but an endothelial transmembrane mediator is still unexplored. The LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) is a transmembrane scavenger receptor that can cause endothelial dysfunction, and its expression is increased in the endothelium of preeclampsia women. In this study, we hypothesized that LOX-1 mediates the effects of preeclampsia-STBEVs on endothelial function. METHODS Preeclampsia-STBEVs were collected by perfusion of placentas from women with preeclampsia and in vitro and ex vivo endothelial cell function were assessed. RESULTS In human umbilical vein endothelial cells, inhibition of LOX-1 with LOX-1 blocking antibody (TS20) reduced the uptake of preeclampsia-STBEVs (61.3±8.8%). TS20 prevented the activation of ERK (extracellular signal-regulated kinase, a kinase downstream of LOX-1) and reduced the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells; 21.1±8.0%) and nitrative stress (23.2±10.3%) that was induced by preeclampsia-STBEVs. Vascular function was assessed by wire myography in isolated mesenteric arteries from pregnant rats that were incubated overnight with preeclampsia-STBEVs±TS20. TS20 prevented endothelium-dependent vasodilation impairment induced by preeclampsia-STBEVs. Nitric oxide contribution to the relaxation was reduced by preeclampsia-STBEVs, which was prevented by TS20. Superoxide dismutase or apocynin, an inhibitor of NOX (nicotinamide adenine dinucleotide phosphate oxidase), restored the impaired endothelium-dependent vasodilation in arteries exposed to preeclampsia-STBEVs. CONCLUSIONS Taken together, our findings demonstrate that LOX-1 mediates the endothelial dysfunction induced by preeclampsia-STBEVs. Our study further expands on the mechanisms that may lead to adverse outcomes in preeclampsia and proposes LOX-1 as a potential target for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Escuela de Medicina sede San Felipe (R.V.-L.), Universidad de Valparaíso, Chile
| | - Ricky Liu
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Departamento de Medicina Interna (T. Sáez), Universidad de Valparaíso, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina (T. Sáez), Universidad de Valparaíso, Chile
| | - Tamara Semeria Maitret
- Department of Laboratory Medicine and Pathology (T.S.M.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tatsuya Sawamura
- Departments of Molecular Pathophysiology and Life Innovation, Shinshu University, Matsumoto, Japan (T. Sawamura)
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
13
|
Nair S, Ormazabal V, Carrion F, Handberg A, McIntyre H, Salomon C. Extracellular vesicle-mediated targeting strategies for long-term health benefits in gestational diabetes. Clin Sci (Lond) 2023; 137:1311-1332. [PMID: 37650554 PMCID: PMC10472199 DOI: 10.1042/cs20220150] [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: 02/20/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Extracellular vesicles (EVs) are critical mediators of cell communication, playing important roles in regulating molecular cross-talk between different metabolic tissues and influencing insulin sensitivity in both healthy and gestational diabetes mellitus (GDM) pregnancies. The ability of EVs to transfer molecular cargo between cells imbues them with potential as therapeutic agents. During pregnancy, the placenta assumes a vital role in metabolic regulation, with multiple mechanisms of placenta-mediated EV cross-talk serving as central components in GDM pathophysiology. This review focuses on the role of the placenta in the pathophysiology of GDM and explores the possibilities and prospects of targeting the placenta to address insulin resistance and placental dysfunction in GDM. Additionally, we propose the use of EVs as a novel method for targeted therapeutics in treating the dysfunctional placenta. The primary aim of this review is to comprehend the current status of EV targeting approaches and assess the potential application of these strategies in placental therapeutics, thereby delivering molecular cargo and improving maternal and fetal outcomes in GDM. We propose that EVs have the potential to revolutionize GDM management, offering hope for enhanced maternal-fetal health outcomes and more effective treatments.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicle in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Australia
| | - Valeska Ormazabal
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepcion, Concepción, Chile
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - H David McIntyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicle in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Australia
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| |
Collapse
|
14
|
Kazatsker MM, Sharabi-Nov A, Meiri H, Sammour R, Sammar M. Augmented Placental Protein 13 in Placental-Associated Extracellular Vesicles in Term and Preterm Preeclampsia Is Further Elevated by Corticosteroids. Int J Mol Sci 2023; 24:12051. [PMID: 37569423 PMCID: PMC10419231 DOI: 10.3390/ijms241512051] [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: 05/29/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Placental protein 13 (PP13) is a regulatory protein involved in remodeling the vascular system of the pregnancy and extending the immune tolerance of the mother to the growing fetus. PP13 is localized on the surface of the syncytiotrophoblast. An ex vivo placental model shows that the PP13 is released via placental-associated extracellular vesicles (PEVs) to the maternal uterine vein. This exploratory study aimed to determine PEV-associated PP13 in the maternal circulation as compared to the known soluble fraction since each has a specific communication pathway. Patients admitted to Bnai Zion Medical Center for delivery were recruited, and included 19 preeclampsia (PE) patients (7 preterm PE gestational age < 37 weeks' gestation), 16 preterm delivery (PTD, delivery at GA < 37 weeks' gestation), and 15 matched term delivery controls. Treatment by corticosteroids (Celestone), which is often given to patients with suspected preterm PE and PTD, was recorded. The PEV proteome was purified from the patients' plasma by size exclusion chromatography (SEC) to separate the soluble and PEV-associated PP13. The total level of PP13 (soluble and PEV-associated) was determined using mild detergent that depleted the PEV proteome. PP13 fractions were determined by ELISA with PP13 specific antibodies. ELISA with alkaline phosphatase (PLAP)- and cluster differentiation 63 (CD63)-specific antibodies served to verify the placental origin of the PEVs. SPSS was used for statistical analysis. The patients' medical, pregnancy, and delivery records in all groups were similar except, as expected, that a larger number of PE and PTD patients had smaller babies who were delivered earlier, and the PE patients had hypertension and proteinuria. The SEC analysis detected the presence of PP13 in the cargo of the PEVs and on their surface, in addition to the known soluble fraction. The median soluble PP13 was not significantly different across the PE, PTD, and term delivery control groups. However, after depleting the PEV of their proteome, the total PP13 (soluble and PEV-associated) was augmented in the cases of preterm PE, reaching 2153 pg/mL [IQR 1866-2838] but not in cases of PTD reaching 1576 pg/mL [1011-2014] or term delivery groups reaching 964 pg/mL [875-1636]), p < 0.01. On the surface of the circulating PEV from PTD patients, there was a decrease in PP13. Corticosteroid treatment was accompanied by a massive depletion of PP13 from the PEV, especially in preterm PE patients. This exploratory study is, thus, the first to determine PEV-associated PP13 in maternal circulation, providing a quantitative determination of the soluble and the PEV-associated fractions, and it shows that the latter is the larger. We found an increase in the amount of PP13 carried via the PEV-associated pathway in PE and PTD patients compared to term delivery cases, which was further augmented when the patients were treated with corticosteroids, especially in preterm PE. The signal conveyed by this novel communication pathway warrants further research to investigate these two differential pathways for the liberation of PP13.
Collapse
Affiliation(s)
- Marina Marks Kazatsker
- Maternal and Fetal Medicine Unit, Department of Obstetrics and Gynecology, Bnai-Zion University Medical Center, Haifa 3498838, Israel; (M.M.K.); (R.S.)
| | - Adi Sharabi-Nov
- Department of Statistics, Tel Hai Academic College, Tel Hai 122103, Israel;
- Department of Statistics, Ziv Medical Center, Safed 131000, Israel
| | - Hamutal Meiri
- Hy-Laboratories Ltd., Rehovot 7670606, Israel;
- TeleMarpe Ltd., 21 Beit El St., Tel Aviv 6908742, Israel
| | - Rami Sammour
- Maternal and Fetal Medicine Unit, Department of Obstetrics and Gynecology, Bnai-Zion University Medical Center, Haifa 3498838, Israel; (M.M.K.); (R.S.)
| | - Marei Sammar
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude Academic College of Engineering, 51 Snunit St., Karmiel 2161002, Israel
| |
Collapse
|
15
|
Nguyen CM, Sallam M, Islam MS, Clack K, Soda N, Nguyen NT, Shiddiky MJA. Placental Exosomes as Biomarkers for Maternal Diseases: Current Advances in Isolation, Characterization, and Detection. ACS Sens 2023. [PMID: 37449399 DOI: 10.1021/acssensors.3c00689] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Serving as the interface between fetal and maternal circulation, the placenta plays a critical role in fetal growth and development. Placental exosomes are small membrane-bound extracellular vesicles released by the placenta during pregnancy. They contain a variety of biomolecules, including lipids, proteins, and nucleic acids, which can potentially be biomarkers of maternal diseases. An increasing number of studies have demonstrated the utility of placental exosomes for the diagnosis and monitoring of pathological conditions such as pre-eclampsia and gestational diabetes. This suggests that placental exosomes may serve as new biomarkers in liquid biopsy analysis. This review provides an overview of the current understanding of the biological function of placental exosomes and their potential as biomarkers of maternal diseases. Additionally, this review highlights current barriers and the way forward for standardization and validation of known techniques for exosome isolation, characterization, and detection. Finally, microfluidic devices for exosome research are discussed.
Collapse
Affiliation(s)
- Cong Minh Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Mohamed Sallam
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Md Sajedul Islam
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Kimberley Clack
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Narshone Soda
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Muhammad J A Shiddiky
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Rural Health Research Institute, Charles Sturt University, Orange, NSW 2800, Australia
| |
Collapse
|
16
|
Miguel AC, Aurora GH, Alejandro SP. Cardiosome-mediated protection in myocardial ischemia. Clin Chim Acta 2023; 545:117374. [PMID: 37150341 DOI: 10.1016/j.cca.2023.117374] [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: 02/20/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/09/2023]
Abstract
Cardiosomes, exosomes released in cardiospheres by cardiomyocytes and progenitor cells, communicate locally and at a distance from different tissues, promoting beneficial cellular changes. For example, miRNAs have emerged as regulators of intercellular communication via transport by extracellular vesicles in general and cardiosomes specifically. Although cardiosomes are considered biomarkers owing to their immense biomedical application in various clinical fields, their role in cardiovascular diseases remains unclear. This mini-review examines the experimental and clinical evidence for cardiosomes as non-invasive diagnostic, treatment and prognostic tools in acute myocardial infarction, the novelty of which is often lost in medical practice. In addition, we discuss the potential role of cardiosomes in physiologic mechanisms and cell signaling in cardiac conditioning strategies against reperfusion injury.
Collapse
Affiliation(s)
- Arroyo-Campuzano Miguel
- Department of Biomedicine Cardiovascular, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Gil-Hernández Aurora
- Department of Biomedicine Cardiovascular, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Silva-Palacios Alejandro
- Department of Biomedicine Cardiovascular, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
17
|
Bueno-Sánchez JC, Gómez-Gutiérrez AM, Maldonado-Estrada JG, Quintana-Castillo JC. Expression of placental glycans and its role in regulating peripheral blood NK cells during preeclampsia: a perspective. Front Endocrinol (Lausanne) 2023; 14:1087845. [PMID: 37206444 PMCID: PMC10190602 DOI: 10.3389/fendo.2023.1087845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/03/2023] [Indexed: 05/21/2023] Open
Abstract
Preeclampsia is a pregnancy-related multisystem disorder characterized by altered trophoblast invasion, oxidative stress, exacerbation of systemic inflammatory response, and endothelial damage. The pathogenesis includes hypertension and mild-to-severe microangiopathy in the kidney, liver, placenta, and brain. The main mechanisms involved in its pathogenesis have been proposed to limit trophoblast invasion and increase the release of extracellular vesicles from the syncytiotrophoblast into the maternal circulation, exacerbating the systemic inflammatory response. The placenta expresses glycans as part of its development and maternal immune tolerance during gestation. The expression profile of glycans at the maternal-fetal interface may play a fundamental role in physiological pregnancy changes and disorders such as preeclampsia. It is unclear whether glycans and their lectin-like receptors are involved in the mechanisms of maternal-fetal recognition by immune cells during pregnancy homeostasis. The expression profile of glycans appears to be altered in hypertensive disorders of pregnancy, which could lead to alterations in the placental microenvironment and vascular endothelium in pregnancy conditions such as preeclampsia. Glycans with immunomodulatory properties at the maternal-fetal interface are altered in early-onset severe preeclampsia, implying that innate immune system components, such as NK cells, exacerbate the systemic inflammatory response observed in preeclampsia. In this article, we discuss the evidence for the role of glycans in gestational physiology and the perspective of glycobiology on the pathophysiology of hypertensive disorders in gestation.
Collapse
Affiliation(s)
- Julio C. Bueno-Sánchez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Department of Obstetrics and Gynecology, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares en Trastornos del Embarazo (RIVATREM), Chillan, Chile
| | - Alejandra M. Gómez-Gutiérrez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Juan G. Maldonado-Estrada
- One Health and Veterinary Innovative Research & Development (OHVRI) Research Group, Escuela de Medicina Veterinaria, Universidad de Antioquia, Medellín, Colombia
| | | |
Collapse
|
18
|
Kandzija N, Rahbar M, Jones GD, Motta-Mejia C, Zhang W, Couch Y, Neuhaus AA, Kishore U, Sutherland BA, Redman C, Vatish M. Placental capillary pericytes release excess extracellular vesicles under hypoxic conditions inducing a pro-angiogenic profile in term pregnancy. Biochem Biophys Res Commun 2023; 651:20-29. [PMID: 36774662 DOI: 10.1016/j.bbrc.2023.02.015] [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: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Pericytes are multifunctional cells wrapped around capillary endothelia, essential for vascular health, development, and blood flow regulation, although their role in human placental chorionic villi has not been fully explored. The second half of normal pregnancy is characterized by a progressive decline in placental and fetal oxygen levels which, by term, comprises a substantial degree of hypoxia. We hypothesized this hypoxia would stimulate pericyte regulation of chorionic villous capillary function. This study's objective was to investigate the role of hypoxia on normal term placental pericytes (PLVP) and their signaling to endothelial cells. First, we confirmed fetoplacental hypoxia at term by a new analysis of umbilical arterial blood oxygen tension of 3,010 healthy singleton neonates sampled at caesarean section and before labor. We then measured the release of cytokines, chemokines, and small extracellular vesicles (PLVPsv), from PLVP cultured at 20%, 8% and 1% O2. As O2 levels decreased, secreted cytokines and chemokines [interleukin-6 (IL-6), interleukin-1α (IL-1α) and vascular endothelial growth factor (VEGF)], and small extracellular vesicle markers, (Alix, Syntenin and CD9) increased significantly in the culture supernatants. When primary human umbilical vein endothelial cells (HUVEC) were cultured with PLVPsv, polygon formation, number, and tube formation length was significantly increased compared to cells not treated with PLVPsv, indicating PLVPsv stimulated angiogenesis. We conclude that adding PLVPsv stimulates angiogenesis and vessel stabilization on neighboring endothelial cells in response to hypoxia in term pregnancy compared to no addition of PLVPsv. Our finding that PLVP can release angiogenic molecules via extracellular vesicles in response to hypoxia may apply to other organ systems.
Collapse
Affiliation(s)
- Neva Kandzija
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Maryam Rahbar
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Gabriel Davis Jones
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Carolina Motta-Mejia
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK; Biosciences Division, College of Health and Life Sciences, Brunel University London, London, UK
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Yvonne Couch
- Acute Stroke Program, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ain A Neuhaus
- Acute Stroke Program, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Uday Kishore
- Biosciences Division, College of Health and Life Sciences, Brunel University London, London, UK
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Christopher Redman
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
19
|
Sokolov D, Gorshkova A, Markova K, Milyutina Y, Pyatygina K, Zementova M, Korenevsky A, Mikhailova V, Selkov S. Natural Killer Cell Derived Microvesicles Affect the Function of Trophoblast Cells. MEMBRANES 2023; 13:213. [PMID: 36837716 PMCID: PMC9963951 DOI: 10.3390/membranes13020213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
The interaction of natural killer (NK) and trophoblast cells underlies the formation of immune tolerance in the mother-fetus system and the maintenance of the physiological course of pregnancy. In addition, NK cells affect the function of trophoblast cells, interacting with them via the receptor apparatus and through the production of cytokines. Microvesicles (MVs) derived from NK cells are able to change the function of target cells. However, in the overall pattern of interactions between NK cells and trophoblasts, the possibility that both can transmit signals to each other via MVs has not been taken into account. Therefore, the aim of this study was to assess the effect of NK cell-derived MVs on the phenotype, proliferation, and migration of trophoblast cells and their expression of intracellular messengers. We carried out assays for the detection of content transferred from MV to trophoblasts. We found that NK cell-derived MVs did not affect the expression of CD54, CD105, CD126, CD130, CD181, CD119, and CD120a receptors in trophoblast cells or lead to the appearance of CD45 and CD56 receptors in the trophoblast membrane. Further, the MVs reduced the proliferation but increased the migration of trophoblasts with no changes to their viability. Incubation of trophoblast cells in the presence of MVs resulted in the activation of STAT3 via pSTAT3(Ser727) but not via pSTAT3(Tyr705). The treatment of trophoblasts with MVs did not result in the phosphorylation of STAT1 and ERK1/2. The obtained data indicate that NK cell-derived MVs influence the function of trophoblast cells, which is accompanied by the activation of STAT3 signaling.
Collapse
|
20
|
Pham QN, Winter M, Milanova V, Young C, Condina MR, Hoffmann P, Pham NTH, Tung TT, Losic D, Thierry B. Magnetic enrichment of immuno-specific extracellular vesicles for mass spectrometry using biofilm-derived iron oxide nanowires. NANOSCALE 2023; 15:1236-1247. [PMID: 36541661 DOI: 10.1039/d2nr05619d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immuno-specific enrichment of extracellular vesicles (EVs) can provide important information into cellular pathways underpinning various pathologies and for non-invasive diagnostics, including mass spectrometry-based analyses. Herein, we report an optimised protocol for immuno-magnetic enrichment of specific EV subtypes and their subsequent processing with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Specifically, we conjugated placental alkaline phosphatase (PLAP) antibodies to magnetic iron oxide nanowires (NWs) derived from bacterial biofilms and demonstrated the utility of this approach by enriching placenta-specific EVs (containing PLAP) from cell culture media. We demonstrate efficient PLAP+ve EV enrichment for both NW-PLAP and Dynabeads™-PLAP, with high PLAP protein recovery (83.7 ± 8.9% and 83.2 ± 5.9%, respectively), high particle-to-protein ratio (7.5 ± 0.7 × 109 and 7.1 ± 1.2 × 109, respectively), and low non-specific binding of non-target EVs (7 ± 3.2% and 5.4 ± 2.2%, respectively). Furthermore, our optimized EV enrichment and processing approach identified 2518 and 2545 protein groups with LC-MS/MS for NW-PLAP and Dynabead™-PLAP, respectively, with excellent reproducibility (Pearson correlation 0.986 and 0.988). These findings demonstrate that naturally occurring iron oxide NWs have comparable performance to current gold standard immune-magnetic beads. The optimized immuno-specific EV enrichment for LC-MS/MS method provides a low-cost and highly-scalable yet efficient, high-throughput approach for quality EV proteomic studies.
Collapse
Affiliation(s)
- Quang Nghia Pham
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Marnie Winter
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Valentina Milanova
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Clifford Young
- Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, South Australia 5000, Australia
| | - Mark R Condina
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Peter Hoffmann
- Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, South Australia 5000, Australia
| | - Nguyen T H Pham
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tran Thanh Tung
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| |
Collapse
|
21
|
Qiu L, Liu X, Zhu L, Luo L, Sun N, Pei R. Current Advances in Technologies for Single Extracellular Vesicle Analysis and Its Clinical Applications in Cancer Diagnosis. BIOSENSORS 2023; 13:129. [PMID: 36671964 PMCID: PMC9856491 DOI: 10.3390/bios13010129] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/31/2022] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) have been regarded as one of the most potential diagnostic biomarkers for different cancers, due to their unique physiological and pathological functions. However, it is still challenging to precisely analyze the contents and sources of EVs, due to their heterogeneity. Herein, we summarize the advances in technologies for a single EV analysis, which may provide new strategies to study the heterogeneity of EVs, as well as their cargo, more specifically. Furthermore, the applications of a single EV analysis on cancer early diagnosis are also discussed.
Collapse
Affiliation(s)
- Lei Qiu
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
- Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xingzhu Liu
- Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Libo Zhu
- Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Liqiang Luo
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Na Sun
- Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
22
|
Menzies FM. Immunology of Pregnancy and Systemic Consequences. Curr Top Microbiol Immunol 2023; 441:253-280. [PMID: 37695432 DOI: 10.1007/978-3-031-35139-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Pregnancy is an immunological paradox, with renowned Nobel Prize winning transplantation biologist Sir Peter Brian Medawar being the first to introduce this concept back in 1953. This concept considers how the maternal immune system can tolerate the developing fetus, which is 50% antigenically foreign to the uterus. There have been significant advances in our understanding of the immune system in regulating fertility, pregnancy and in complications of these, and what was once considered a paradox can be seen as a highly evolved system. Indeed, the complexity of the maternal-fetal interface along with our ever-advancing knowledge of immune cells and mediators means that we have a better understanding of these interactions, with gaps still present. This chapter will summarise the key aspects of the role of the immune system at each stage of pregnancy and highlight the recent advances in our knowledge.
Collapse
Affiliation(s)
- Fiona M Menzies
- School of Health and Life Sciences, University of the West of Scotland, Lanarkshire, UK.
| |
Collapse
|
23
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Quon A, Wong M, Pink D, Lewis J, Vatish M, Davidge S, Cooke CL. Placenta-derived extracellular vesicles from preeclamptic and healthy pregnancies impair ex vivo vascular endothelial function. Biosci Rep 2022; 42:BSR20222185. [PMID: 36408626 PMCID: PMC9760606 DOI: 10.1042/bsr20222185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy syndrome characterized by new-onset hypertension and end-organ dysfunction. The pathophysiology of PE remains undetermined, but it is thought that maternal vascular dysfunction plays a central role, potentially due, in part, to the release of syncytiotrophoblast-derived extracellular vesicles (STBEVs) into the maternal circulation by a dysfunctional placenta. STBEVs from normal pregnancies (NP) impair vascular function, but the effect of PE STBEVs (known to differ in composition with elevated circulating levels) on vascular function are not known. We hypothesized that PE STBEVs have more detrimental effects on vascular function compared with NP STBEVs. STBEVs were collected by perfusion of placentas from women with NP or PE. Mesenteric arteries from pregnant rats were incubated overnight with NP or PE STBEVs, and vascular function was assessed by wire myography. NP and PE STBEVs impaired endothelial function, partially by reducing nitric oxide (NO) bioavailability. Incubation of human umbilical vein endothelial cells with NP and PE STBEVs increased nuclear factor κ-light-chain-enhancer of activated B cell (NF-κB) activation, reactive oxygen species, nitrotyrosine levels, and reduced NO levels. However, PE STBEVs increased NF-κB activation and nitrotyrosine levels to a lesser extent than NP STBEVs. Taken together, no greater impact of PE STBEVs compared with NP STBEVs on endothelial function was found. However, the impaired vascular function by PE STBEVs and increased levels of STBEVs in PE suggest PE STBEVs may contribute to maternal vascular dysfunction in PE. Our study further expands on the potential mechanisms that lead to adverse outcomes in PE and provides potential targets for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| | - Ricky Liu
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
- Department of Physiology, Edmonton, Alberta, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| | - Michael Wong
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Nanostics Inc., Edmonton, Alberta, Canada
| | - Desmond Pink
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Nanostics Inc., Edmonton, Alberta, Canada
| | - John Lewis
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Nanostics Inc., Edmonton, Alberta, Canada
| | - Manu Vatish
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, U.K
| | - Sandra T. Davidge
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
- Department of Physiology, Edmonton, Alberta, Canada
| | - Christy-Lynn M. Cooke
- Department of Obstetrics and Gynecology, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Ortega MA, Fraile-Martínez O, García-Montero C, Paradela A, Asunción Sánchez-Gil M, Rodriguez-Martin S, De León-Luis JA, Pereda-Cerquella C, Bujan J, Guijarro LG, Alvarez-Mon M, García-Honduvilla N. Unfolding the role of placental-derived Extracellular Vesicles in Pregnancy: From homeostasis to pathophysiology. Front Cell Dev Biol 2022; 10:1060850. [PMID: 36478738 PMCID: PMC9720121 DOI: 10.3389/fcell.2022.1060850] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/02/2022] [Indexed: 08/11/2023] Open
Abstract
The human placenta is a critical structure with multiple roles in pregnancy, including fetal nutrition and support, immunological, mechanical and chemical barrier as well as an endocrine activity. Besides, a growing body of evidence highlight the relevance of this organ on the maternofetal wellbeing not only during gestation, but also from birth onwards. Extracellular vesicles (EVs) are complex macromolecular structures of different size and content, acting as carriers of a diverse set of molecules and information from donor to recipient cells. Since its early development, the production and function of placental-derived EVs are essential to ensure an adequate progress of pregnancy. In turn, the fetus receives and produce their own EVs, highlighting the importance of these components in the maternofetal communication. Moreover, several studies have shown the clinical relevance of EVs in different obstetric pathologies such as preeclampsia, infectious diseases or gestational diabetes, among others, suggesting that they could be used as pathophysiological biomarkers of these diseases. Overall, the aim of this article is to present an updated review of the published basic and translational knowledge focusing on the role of placental-derived EVs in normal and pathological pregnancies. We suggest as well future lines of research to take in this novel and promising field.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | | | - María Asunción Sánchez-Gil
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| | - Sonia Rodriguez-Martin
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Service of Pediatric, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Juan A. De León-Luis
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Ma-drid, Madrid, Spain
| | - Claude Pereda-Cerquella
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Luis G. Guijarro
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), Department of System Biology, University of Alcalá, Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), University Hospital Príncipe de Asturias, Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| |
Collapse
|
25
|
Smolkova B, Kataki A, Earl J, Ruz-Caracuel I, Cihova M, Urbanova M, Buocikova V, Tamargo S, Rovite V, Niedra H, Schrader J, Kohl Y. Liquid biopsy and preclinical tools for advancing diagnosis and treatment of patients with pancreatic neuroendocrine neoplasms. Crit Rev Oncol Hematol 2022; 180:103865. [DOI: 10.1016/j.critrevonc.2022.103865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
|
26
|
Tracking matricellular protein SPARC in extracellular vesicles as a non-destructive method to evaluate lipid-based antifibrotic treatments. Commun Biol 2022; 5:1155. [PMID: 36310239 PMCID: PMC9618575 DOI: 10.1038/s42003-022-04123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Uncovering the complex cellular mechanisms underlying hepatic fibrogenesis could expedite the development of effective treatments and noninvasive diagnosis for liver fibrosis. The biochemical complexity of extracellular vesicles (EVs) and their role in intercellular communication make them an attractive tool to look for biomarkers as potential alternative to liver biopsies. We developed a solid set of methods to isolate and characterize EVs from differently treated human hepatic stellate cell (HSC) line LX-2, and we investigated their biological effect onto naïve LX-2, proving that EVs do play an active role in fibrogenesis. We mined our proteomic data for EV-associated proteins whose expression correlated with HSC treatment, choosing the matricellular protein SPARC as proof-of-concept for the feasibility of fluorescence nanoparticle-tracking analysis to determine an EV-based HSCs’ fibrogenic phenotype. We thus used EVs to directly evaluate the efficacy of treatment with S80, a polyenylphosphatidylcholines-rich lipid, finding that S80 reduces the relative presence of SPARC-positive EVs. Here we correlated the cellular response to lipid-based antifibrotic treatment to the relative presence of a candidate protein marker associated with the released EVs. Along with providing insights into polyenylphosphatidylcholines treatments, our findings pave the way for precise and less invasive diagnostic analyses of hepatic fibrogenesis. A method is developed to isolate and characterize extracellular vesicles (EVs) from human hepatic stellate cells and proteomics reveals that the matricellular protein SPARC may be used as an EV marker after lipid-based antifibrotic treatment.
Collapse
|
27
|
Kwon Y, Park J. Methods to analyze extracellular vesicles at single particle level. MICRO AND NANO SYSTEMS LETTERS 2022. [DOI: 10.1186/s40486-022-00156-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractExtracellular vesicles (EVs) are nano-sized vesicles derived from cells that transport biomaterials between cells through biofluids. Due to their biological role and components, they are considered as potential drug carriers and for diagnostic applications. Today's advanced nanotechnology enables single-particle-level analysis that was difficult in the past due to its small size below the diffraction limit. Single EV analysis reveals the heterogeneity of EVs, which could not be discovered by various ensemble analysis methods. Understanding the characteristics of single EVs enables more advanced pathological and biological researches. This review focuses on the advanced techniques employed for EV analysis at the single particle level and describes the principles of each technique.
Collapse
|
28
|
Macrophage-Derived Exosomes in TLR9-/- Mice Ameliorate Sepsis-Induced Mitochondrial Oxidative Stress and Apoptosis in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5719974. [PMID: 36225174 PMCID: PMC9550441 DOI: 10.1155/2022/5719974] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 12/29/2022]
Abstract
Objective To explore the mechanisms of TLR9 from macrophages on mitochondrial apoptosis in cardiomyocytes at early stage of sepsis. Methods The in vivo and in vitro sepsis mice were bone marrow transplantation (BMT) with wild type (WT) or (toll-like receptor 9) TLR9 knockout (−/− or KO) myeloid cells and then constructed by cecum ligation and puncture (CLP) as vivo experiment and cardiomyocytes cocultured with WT or TLR9-deficient macrophages treated with LPS as vitro experiment, respectively. Sepsis model were performed by CLP. The expression levels of exosome, PI3K/AKT, and ERK1/2, inflammatory factors, and apoptotic proteins were tested by western blot in vivo. Besides, associated apoptotic proteins and JC-1 fluorescence assay were tested in vitro. Results The expressions of p-PI3K, p-AKT, exosome markers (CD9, CD63, and TSG101), p-ERK1/2, TNF-α, IFN-γ, IL-1β, and cleaved-caspase-3/-9 were significantly increased in septic mice vs. control mice, and these proteins were declined dramatically in TLR9−/− BMT mice vs. WT BMT mice in sepsis mice models. Meanwhile, the protein expression of cytochrome C, cleaved-caspase-3, and cleaved-caspase-9 increased significantly in primary mouse myocardial cells cocultured with TLR9−/− or WT macrophages stimulated with LPS, and these mitochondrial apoptotic proteins as well as the green 5,5',6,6'-tetrachloro-1,1',3,3'- tetraethylbenzimidazolcarbocyanine iodide (JC-1) fluorescence were dramatically lower in LPS-stimulated cardiomyocytes cocultured with TLR9−/− than with WT macrophages. Conclusion TLR9−/− in macrophages suppressed the inflammatory reaction as well as the exosome secretion and resulted in the inhibition of apoptosis and oxidative stress in sepsis-induced cardiomyopathy.
Collapse
|
29
|
Sheldon H, Zhang W, Bridges E, Ang KH, Lin S, Masiero M, Li D, Handford PA, Whiteman P, Fischer R, Buffa F, Vatish M, Banham AH, Harris AL. ELTD1 is present in extracellular vesicles derived from endothelial cells as a cleaved extracellular domain which induces in vivo angiogenesis. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e52. [PMID: 38939053 PMCID: PMC11080856 DOI: 10.1002/jex2.52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 06/29/2024]
Abstract
ELTD1/ADGRL4 is an adhesion GPCR with an important role in angiogenesis. We recently identified a role for ELTD1 in wound repair and inflammation. Activation of ELTD1 in endothelial cells results in a type II EMT to myofibroblast-like cells that have enhanced angiogenic ability. Furthermore, expression of Eltd1 in murine breast cancer cells increases tumour growth by increasing blood vessel size and perfusion and by creating an immunosuppressive microenvironment. As extracellular vesicles (EVs) are known to be involved in vascular development, growth and maturation we investigated the composition and functional effects of the EVs isolated from ELTD1 expressing cells to elucidate their role in these processes. A highly glycosylated form of the extracellular domain (ECD) of ELTD1 is readily incorporated into EVs. Using mass spectrometry-based proteomics we identified proteins that are enriched in ELTD1-EVs and are involved in haemostasis and immune responses. ELTD1 enriched EVs were pro-angiogenic in vivo and in vitro and the presence of the ECD alone induced endothelial sprouting. In endothelial cells experiencing laminar flow, ELTD1 levels were reduced in the EVs when they are quiescent, showing a relationship between ELTD1 and the activation state of the endothelium. Using FACS, we detected a significant increase in vesicular ELTD1 in the plasma of patients with preeclampsia, a condition characterized by endothelial dysfunction. These data confirm a role for ELTD1 in wound repair and inflammation and reveal its potential as a biomarker of vessel dysfunction.
Collapse
Affiliation(s)
- Helen Sheldon
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Esther Bridges
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Koon Hwee Ang
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Salwa Lin
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Massimo Masiero
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Demin Li
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | | | - Pat Whiteman
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Roman Fischer
- Nuffield Department of MedicineTarget Discovery InstituteOxford University, NDM Research BuildingOxfordUK
| | - Francesca Buffa
- Department of OncologyUniversity of OxfordChurchill HospitalOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Adrian L. Harris
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| |
Collapse
|
30
|
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
|
31
|
Babaker MA, Aljoud FA, Alkhilaiwi F, Algarni A, Ahmed A, Khan MI, Saadeldin IM, Alzahrani FA. The Therapeutic Potential of Milk Extracellular Vesicles on Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23126812. [PMID: 35743255 PMCID: PMC9224713 DOI: 10.3390/ijms23126812] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer remains one of the leading prevalent cancers in the world and is the fourth most common cause of death from cancer. Unfortunately, the currently utilized chemotherapies fail in selectively targeting cancer cells and cause harm to healthy cells, which results in profound side effects. Researchers are focused on developing anti-cancer targeted medications, which is essential to making them safer, more effective, and more selective and to maximizing their therapeutic benefits. Milk-derived extracellular vesicles (EVs) from camels and cows have attracted much attention as a natural substitute product that effectively suppresses a wide range of tumor cells. This review sheds light on the biogenesis, methods of isolation, characterization, and molecular composition of milk EVs as well as the therapeutic potentials of milk EVs on colorectal cancer.
Collapse
Affiliation(s)
- Manal A. Babaker
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Chemistry, Faculty of Science, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Fadwa A. Aljoud
- Regenerative Medicine Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.); (F.A.)
| | - Faris Alkhilaiwi
- Regenerative Medicine Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.); (F.A.)
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulrahman Algarni
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 73221, Saudi Arabia;
| | - Asif Ahmed
- MirZyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Birmingham B7 4BB, UK;
- School of Health Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Mohammad Imran Khan
- Centre of Artificial Intelligence in Precision Medicines (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Islam M. Saadeldin
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Correspondence: (I.M.S.); (F.A.A.)
| | - Faisal A. Alzahrani
- MirZyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Birmingham B7 4BB, UK;
- Centre of Artificial Intelligence in Precision Medicines (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Embryonic Stem Cells Unit, Department of Biochemistry, Faculty of Science, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (I.M.S.); (F.A.A.)
| |
Collapse
|
32
|
Ali Moussa HY, Manaph N, Ali G, Maacha S, Shin KC, Ltaief SM, Gupta V, Tong Y, Ponraj J, Salloum-Asfar S, Mansour S, Al-Shaban FA, Kim HG, Stanton LW, Grivel JC, Abdulla SA, Al-Shammari AR, Park Y. Single Extracellular Vesicle Analysis Using Flow Cytometry for Neurological Disorder Biomarkers. Front Integr Neurosci 2022; 16:879832. [PMID: 35655952 PMCID: PMC9152125 DOI: 10.3389/fnint.2022.879832] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles released from cells to the extracellular space, involved in cell-to-cell communication by the horizontal transfer of biomolecules such as proteins and RNA. Because EVs can cross the blood-brain barrier (BBB), circulating through the bloodstream and reflecting the cell of origin in terms of disease prognosis and severity, the contents of plasma EVs provide non-invasive biomarkers for neurological disorders. However, neuronal EV markers in blood plasma remain unclear. EVs are very heterogeneous in size and contents, thus bulk analyses of heterogeneous plasma EVs using Western blot and ELISA have limited utility. In this study, using flow cytometry to analyze individual neuronal EVs, we show that our plasma EVs isolated by size exclusion chromatography are mainly CD63-positive exosomes of endosomal origin. As a neuronal EV marker, neural cell adhesion molecule (NCAM) is highly enriched in EVs released from induced pluripotent stem cells (iPSCs)-derived cortical neurons and brain organoids. We identified the subpopulations of plasma EVs that contain NCAM using flow cytometry-based individual EV analysis. Our results suggest that plasma NCAM-positive neuronal EVs can be used to discover biomarkers for neurological disorders.
Collapse
Affiliation(s)
- Houda Yasmine Ali Moussa
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Nimshitha Manaph
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Gowher Ali
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Selma Maacha
- Deep Phenotyping Core, Research Branch, Sidra Medicine, Doha, Qatar
| | - Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Samia M. Ltaief
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Vijay Gupta
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Yongfeng Tong
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Janarthanan Ponraj
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Said Mansour
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Fouad A. Al-Shaban
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Lawrence W. Stanton
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | | | - Sara A. Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Abeer R. Al-Shammari
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- Abeer R. Al-Shammari,
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- *Correspondence: Yongsoo Park,
| |
Collapse
|
33
|
Ding Z, Lu Y, Wei Y, Song D, Xu Z, Fang J. DNA-Engineered iron-based metal-organic framework bio-interface for rapid visual determination of exosomes. J Colloid Interface Sci 2022; 612:424-433. [PMID: 34999547 DOI: 10.1016/j.jcis.2021.12.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/28/2022]
Abstract
In this study, a rapid, low-cost and facile method for detecting exosomes was developed by engineering DNA ligands on the surface of an iron-based metal-organic framework (Fe-MOF). Aptamers of exosomal transmembrane CD63 protein (CD63-aptamers) were utilized as both the optically active layer and the exosome-specific recognition element to engineer an Fe-MOF bio-interface for high-efficiency regulation of the catalytic behavior of Fe-MOF toward the chromogenic substrate. The effective enhancement of the intrinsic peroxidase-like catalytic activity was confirmed via the self-assembly of CD63-aptamers on the surface of Fe-MOF. The specific binding of exosomes with CD63-aptamers altered the conformation of DNA ligands on the surface of Fe-MOF, contributing to sensitive variation in Fe-MOF catalytic activity. This directly produced a distinct color change and enabled the visual detection of exosomes. Via one-step "mixing-and-detection", the Fe-MOF bio-interface exhibited excellent performance in quantitative analysis of exosomes derived from human breast cancer cell lines ranging from 1.1 × 105 to 2.2 × 107 particles/μL with a detection limit of 5.2 × 104 particles/μL. The expression of exosomal CD63 proteins originated from three types of cancer cell lines, including breast cancer, gastric cancer and lung cancer cell lines, was differentiated within only 17 min. Furthermore, the method was successfully applied to the identification of exosomes in serum samples, suggesting its potential in clinical analysis as a valuable tool for the rapid, convenient and economical testing of exosomes.
Collapse
Affiliation(s)
- Ziling Ding
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Yanbing Lu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 110122 Shenyang, China
| | - Yunyun Wei
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Dan Song
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China.
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 110122 Shenyang, China
| |
Collapse
|
34
|
Morelli AE, Sadovsky Y. Extracellular vesicles and immune response during pregnancy: A balancing act. Immunol Rev 2022; 308:105-122. [PMID: 35199366 DOI: 10.1111/imr.13074] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
The mechanisms underlying maternal tolerance of the semi- or fully-allogeneic fetus are intensely investigated. Across gestation, feto-placental antigens interact with the maternal immune system locally within the trophoblast-decidual interface and distantly through shed cells and soluble molecules that interact with maternal secondary lymphoid tissues. The discovery of extracellular vesicles (EVs) as local or systemic carriers of antigens and immune-regulatory molecules has added a new dimension to our understanding of immune modulation prior to implantation, during trophoblast invasion, and throughout the course of pregnancy. New data on immune-regulatory molecules, located on EVs or within their cargo, suggest a role for EVs in negotiating immune tolerance during gestation. Lessons from the field of transplant immunology also shed light on possible interactions between feto-placentally derived EVs and maternal lymphoid tissues. These insights illuminate a potential role for EVs in major obstetrical disorders. This review provides updated information on intensely studied, pregnancy-related EVs, their cargo molecules, and patterns of fetal-placental-maternal trafficking, highlighting potential immune pathways that might underlie immune suppression or activation in gestational health and disease. Our summary also underscores the likely need to broaden the definition of the maternal-fetal interface to systemic maternal immune tissues that might interact with circulating EVs.
Collapse
Affiliation(s)
- Adrian E Morelli
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
35
|
Assessing hypoxic damage to placental trophoblasts by measuring membrane viscosity of extracellular vesicles. Placenta 2022; 121:14-22. [PMID: 35245720 PMCID: PMC9010367 DOI: 10.1016/j.placenta.2022.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/06/2022] [Accepted: 02/22/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION As highly sophisticated intercellular communication vehicles in biological systems, extracellular vesicles (EVs) have been investigated as both promising liquid biopsy-based disease biomarkers and drug delivery carriers. Despite tremendous progress in understanding their biological and physiological functions, mechanical characterization of these nanoscale entities remains challenging due to the limited availability of proper techniques. Especially, whether damage to parental cells can be reflected by the mechanical properties of their EVs remains unknown. METHODS In this study, we characterized membrane viscosities of different types of EVs collected from primary human trophoblasts (PHTs), including apoptotic bodies, microvesicles and small extracellular vesicles, using fluorescence lifetime imaging microscopy (FLIM). The biochemical origin of EV membrane viscosity was examined by analyzing their phospholipid composition, using mass spectrometry. RESULTS We found that different EV types derived from the same cell type exhibit different membrane viscosities. The measured membrane viscosity values are well supported by the lipidomic analysis of the phospholipid compositions. We further demonstrate that the membrane viscosity of microvesicles can faithfully reveal hypoxic injury of the human trophoblasts. More specifically, the membrane of PHT microvesicles released under hypoxic condition is less viscous than its counterpart under standard culture condition, which is supported by the reduction in the phosphatidylethanolamine-to-phosphatidylcholine ratio in PHT microvesicles. DISCUSSION Our study suggests that biophysical properties of released trophoblastic microvesicles can reflect cell health. Characterizing EV's membrane viscosity may pave the way for the development of new EV-based clinical applications.
Collapse
|
36
|
Fokina V, Patrikeeva S, Wang X, Shah M, Shah P, Russell WK, Ahmed MS, Rytting E, Nanovskaya T. Physicochemical and Biological Properties of Membrane Vesicles Derived from Human Term Placentas. J Biomed Nanotechnol 2022; 18:589-599. [PMID: 35484740 DOI: 10.1166/jbn.2022.3255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The purpose of this study was to conduct initial characterization of membrane vesicles isolated from human placenta by agitation of villous tissue (apical and basal) as well as vesicles obtained following dual perfusion of placental lobule. The morphology, physical and biological properties of the isolated vesicles were determined by electron microscopy, dynamic light scattering, and immunoblotting as well as nanoflow liquid chromatography-mass spectrometry proteomics analysis. CD-1 male mice were used to test the biocompatibility of the vesicles in vivo and assess the biodistribution of fluorescently labeled apical and perfusion vesicles. The vesicles obtained following placental perfusion and the apical vesicles had Z-average diameters of 199±23 nm and 246±24 nm, respectively, and demonstrated nanocarrier stability, low toxicity, and low immunogenicity. On the other hand, administration of basal vesicles resulted in animal demise with LD50 of 0.85 μgprotein/g. Both fluorescently labeled apical and perfusion vesicles were detected in the lungs, liver, kidneys, and spleen of CD-1 mice within 24 h of administration. However, there were differences in organ distribution of these vesicles over 24 hours time period. These data suggest that placental apical and perfusion vesicles have a potential for further development as biological vehicles for drug delivery.
Collapse
Affiliation(s)
- Valentina Fokina
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Svetlana Patrikeeva
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiaoming Wang
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mansi Shah
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Poonam Shah
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Erik Rytting
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tatiana Nanovskaya
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
37
|
Lai A, Palma C, Salas A, Carrion F, Salomon C. Targeted Mass Spectrometry-Based Proteomics Method to Quantify Placental Extracellular Vesicles. Methods Mol Biol 2022; 2504:79-89. [PMID: 35467280 DOI: 10.1007/978-1-0716-2341-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Extracellular vesicles (EVs) carry a wide range of molecules, such as proteins, RNAs, and DNA. EVs are secreted from a wide range of cells, including placental cells. Interestingly, EVs secreted from placental cells have been identified in maternal circulation as early as 6 weeks of gestation, and their concentration increases with the gestational age. While there is growing interest in elucidating the role of exosomes during normal and complicated pregnancies, progress in the field has been delayed because of the inability to quantify placental EVs from the maternal circulation. Recent reports have demonstrated the presence of placental-type alkaline phosphatase (PLAP) EVs only in the blood of pregnant women, indicating that PLAP is a marker to identify EVs secreted from the placenta. Therefore, here we describe a workflow to quantify placental EVs from maternal circulation using a targeted proteomics approach based on selecting specific peptides identified in the PLAP protein.
Collapse
Affiliation(s)
- Andrew Lai
- Exosome Biology Laboratory, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Carlos Palma
- Exosome Biology Laboratory, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Alexis Salas
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Carlos Salomon
- Exosome Biology Laboratory, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, Australia.
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
38
|
Gupta S, Mazumder P. Exosomes as diagnostic tools. Adv Clin Chem 2022; 110:117-144. [DOI: 10.1016/bs.acc.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Tersigni C, Furqan Bari M, Cai S, Zhang W, Kandzija N, Buchan A, Miranda F, Di Simone N, Redman CW, Bastie C, Vatish M. Syncytiotrophoblast-derived extracellular vesicles carry apolipoprotein-E and affect lipid synthesis of liver cells in vitro. J Cell Mol Med 2021; 26:123-132. [PMID: 34894055 PMCID: PMC8742183 DOI: 10.1111/jcmm.17056] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
In normal pregnancy, hepatic metabolism adaptation occurs with an increase in lipid biosynthesis. Placental shedding of syncytiotrophoblast‐derived extracellular vesicles (STBEVs) into the maternal circulation constitutes a major signalling mechanism between foetus and mother. We investigated whether STBEVs from normal pregnant women might target liver cells in vitro and induce changes in lipid synthesis. This study was performed at the Nuffield Department of Women's & Reproductive Health, Oxford, UK. STBEVs were obtained by dual‐lobe placental perfusion from 11 normal pregnancies at term. Medium/large and small STBEVs were collected by ultracentrifugation at 10,000g and 150,000g, respectively. STBEVs were analysed by Western blot analysis and flow cytometry for co‐expression of apolipoprotein‐E (apoE) and placental alkaline phosphatase (PLAP). The uptake of STBEVs by liver cells and the effect on lipid metabolism was evaluated using a hepatocarcinoma cell line (HepG2 cells). Data were analysed by one‐way ANOVA and Student's t test. We demonstrated that: (a) STBEVs carry apoE; (b) HepG2 cells take up STBEVs through an apoE‐LDL receptor interaction; (c) STBEV incorporation into HepG2 cells resulted in (i) increased cholesterol release (ELISA); (ii) increased expression of the genes SQLE and FDPS (microarray) involved in cholesterol biosynthesis; (iii) downregulation of the CLOCK gene (microarray and PCR), involved in the circadian negative control of lipid synthesis in liver cells. In conclusion, the placenta may orchestrate the metabolic adaptation of the maternal liver through release of apoE‐positive STBEVs, by increasing lipid synthesis in a circadian‐independent fashion, meeting the nutritional needs of the growing foetus.
Collapse
Affiliation(s)
- Chiara Tersigni
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Dipartimento di Scienze della Salute della Donna, Fondazione Policlinico Universitario A. Gemelli IRCCS, del Bambino e di Sanità Pubblica, Rome, Italy
| | | | - Shijei Cai
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Neva Kandzija
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Alice Buchan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Fabrizio Miranda
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.,Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy
| | - Christopher W Redman
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Claire Bastie
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
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
|
41
|
Bergamelli M, Martin H, Bénard M, Ausseil J, Mansuy JM, Hurbain I, Mouysset M, Groussolles M, Cartron G, Tanguy le Gac Y, Moinard N, Suberbielle E, Izopet J, Tscherning C, Raposo G, Gonzalez-Dunia D, D'Angelo G, Malnou CE. Human Cytomegalovirus Infection Changes the Pattern of Surface Markers of Small Extracellular Vesicles Isolated From First Trimester Placental Long-Term Histocultures. Front Cell Dev Biol 2021; 9:689122. [PMID: 34568315 PMCID: PMC8461063 DOI: 10.3389/fcell.2021.689122] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have increasingly been recognized as key players in a wide variety of physiological and pathological contexts, including during pregnancy. Notably, EVs appear both as possible biomarkers and as mediators involved in the communication of the placenta with the maternal and fetal sides. A better understanding of the physiological and pathological roles of EVs strongly depends on the development of adequate and reliable study models, specifically at the beginning of pregnancy where many adverse pregnancy outcomes have their origin. In this study, we describe the isolation of small EVs from a histoculture model of first trimester placental explants in normal conditions as well as upon infection by human cytomegalovirus. Using bead-based multiplex cytometry and electron microscopy combined with biochemical approaches, we characterized these small EVs and defined their associated markers and ultrastructure. We observed that infection led to changes in the expression level of several surface markers, without affecting the secretion and integrity of small EVs. Our findings lay the foundation for studying the functional role of EVs during early pregnancy, along with the identification of new predictive biomarkers for the severity and outcome of this congenital infection, which are still sorely lacking.
Collapse
Affiliation(s)
- Mathilde Bergamelli
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Hélène Martin
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Mélinda Bénard
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France.,Service de Néonatalogie, CHU Toulouse, Hôpital des Enfants, Toulouse, France
| | - Jérôme Ausseil
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France.,Laboratoire de Biochimie, CHU Toulouse, Hôpital Rangueil, Toulouse, France
| | - Jean-Michel Mansuy
- Laboratoire de Virologie, CHU Toulouse, Hôpital Purpan, Toulouse, France
| | - Ilse Hurbain
- CNRS UMR 144, Structure et Compartiments Membranaires, Institut Curie, Université Paris Sciences et Lettres, Paris, France.,CNRS UMR 144, Plateforme d'Imagerie Cellulaire et Tissulaire (PICT-IBiSA), Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - Maïlys Mouysset
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Marion Groussolles
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France.,Service de Diagnostic Prénatal, CHU Toulouse, Hôpital Paule de Viguier, Toulouse, France.,INSERM UMR 1027, UPS, Equipe SPHERE Epidémiologie et Analyses en Santé Publique: Risques, Maladies Chroniques et Handicaps, Université de Toulouse, Toulouse, France
| | - Géraldine Cartron
- Service de Gynécologie Obstétrique, CHU Toulouse, Hôpital Paule de Viguier, Toulouse, France
| | - Yann Tanguy le Gac
- Service de Gynécologie Obstétrique, CHU Toulouse, Hôpital Paule de Viguier, Toulouse, France
| | - Nathalie Moinard
- Développement Embryonnaire, Fertilité, Environnement (DEFE), INSERM UMR 1203, Université de Toulouse et Université de Montpellier, Montpellier, France.,CECOS, Groupe d'Activité de Médecine de la Reproduction, CHU Toulouse, Hôpital Paule de Viguier, Toulouse, France
| | - Elsa Suberbielle
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Jacques Izopet
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France.,Laboratoire de Virologie, CHU Toulouse, Hôpital Purpan, Toulouse, France
| | - Charlotte Tscherning
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Graça Raposo
- CNRS UMR 144, Structure et Compartiments Membranaires, Institut Curie, Université Paris Sciences et Lettres, Paris, France.,CNRS UMR 144, Plateforme d'Imagerie Cellulaire et Tissulaire (PICT-IBiSA), Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - Daniel Gonzalez-Dunia
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Gisela D'Angelo
- CNRS UMR 144, Structure et Compartiments Membranaires, Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - Cécile E Malnou
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| |
Collapse
|
42
|
Deng Y, Sun Z, Wang L, Wang M, Yang J, Li G. Biosensor-based assay of exosome biomarker for early diagnosis of cancer. Front Med 2021; 16:157-175. [PMID: 34570311 DOI: 10.1007/s11684-021-0884-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022]
Abstract
Cancer imposes a severe threat to people's health and lives, thus pressing a huge medical and economic burden on individuals and communities. Therefore, early diagnosis of cancer is indispensable in the timely prevention and effective treatment for patients. Exosome has recently become an attractive cancer biomarker in noninvasive early diagnosis because of the unique physiology and pathology functions, which reflects remarkable information regarding the cancer microenvironment, and plays an important role in the occurrence and evolution of cancer. Meanwhile, biosensors have gained great attention for the detection of exosomes due to their superior properties, such as convenient operation, real-time readout, high sensitivity, and remarkable specificity, suggesting promising biomedical applications in the early diagnosis of cancer. In this review, the latest advances of biosensors regarding the assay of exosomes were summarized, and the superiorities of exosomes as markers for the early diagnosis of cancer were evaluated. Moreover, the recent challenges and further opportunities of developing effective biosensors for the early diagnosis of cancer were discussed.
Collapse
Affiliation(s)
- Ying Deng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Zhaowei Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Lei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Minghui Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
43
|
Rouillard ME, Sutter PA, Durham OR, Willis CM, Crocker SJ. Astrocyte-Derived Extracellular Vesicles (ADEVs): Deciphering their Influences in Aging. Aging Dis 2021; 12:1462-1475. [PMID: 34527422 PMCID: PMC8407882 DOI: 10.14336/ad.2021.0608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are an abundant and dynamic glial cell exclusive to the central nervous system (CNS). In the context of injury, inflammation, and/or diseases of the nervous system, astrocyte responses, termed reactive astrogliosis, are a recognized pathological feature across a range of conditions and diseases. However, the impact of reactive astrogliosis is not uniform and varies by context and duration (time). In recent years, extracellular communication between glial cells via extracellular vesicles (EVs) has garnered interest as a process connected with reactive astrogliosis. In this review, we relate recent findings on astrocyte-derived extracellular vesicles (ADEVs) with a focus on factors that can influence the effects of ADEVs and identified age related changes in the function of ADEVs. Additionally, we will discuss the current limitations of existing experimental approaches and identify questions that highlight areas for growth in this field, which will continue to enhance our understanding of ADEVs in age-associated processes.
Collapse
Affiliation(s)
- Megan E Rouillard
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Pearl A Sutter
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Olivia R Durham
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Cory M Willis
- 2Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Stephen J Crocker
- 1Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| |
Collapse
|
44
|
Ayala-Ramírez P, Machuca-Acevedo C, Gámez T, Quijano S, Barreto A, Silva JL, Olaya-C M, García-Robles R. Assessment of Placental Extracellular Vesicles-Associated Fas Ligand and TNF-Related Apoptosis-Inducing Ligand in Pregnancies Complicated by Early and Late Onset Preeclampsia. Front Physiol 2021; 12:708824. [PMID: 34366896 PMCID: PMC8342945 DOI: 10.3389/fphys.2021.708824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder that affects 2-8% of pregnancies and is one of the main causes of fetal, neonatal, and maternal mortality and morbidity worldwide. Although PE etiology and pathophysiology remain unknown, there is evidence that the hyperactivation of maternal immunity cells against placental cells triggers trophoblast cell apoptosis and death. It has also been reported that placenta-derived extracellular vesicles (EV) carry Fas ligand (FasL) and Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and trigger apoptosis in Jurkat T cells. This study aimed to quantify and compare FasL and TRAIL expression in EV derived from cultures of placenta explants from women with PE (early versus late) and women with uncomplicated pregnancies. Also, the study assessed EV capacity to induce apoptosis in Jurkat T cells. The authors isolated EV from placenta explant cultures, quantified FasL and TRAIL using ELISA, and analyzed EV apoptosis-inducing capability by flow cytometry. Results showed increased FasL and TRAIL in EV derived from placenta of women with PE, and increased EV apoptosis-inducing capability in Jurkat T cells. These results offer supporting evidence that EV FasL and TRAIL play a role in the pathophysiology of PE.
Collapse
Affiliation(s)
- Paola Ayala-Ramírez
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Catalina Machuca-Acevedo
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Tatiana Gámez
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jaime L. Silva
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Mercedes Olaya-C
- Department of Pathology, Faculty of Medicine, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Reggie García-Robles
- Department of Physiological Sciences, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
45
|
Desterke C, Dang J, Lorenzo HK, Candelier JJ. Roles of tetraspanins during trophoblast development: bioinformatics and new perspectives. Cell Tissue Res 2021; 386:157-171. [PMID: 34278518 DOI: 10.1007/s00441-021-03502-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/28/2021] [Indexed: 11/24/2022]
Abstract
Tetraspanins are a superfamily of membrane proteins found in all eukaryotic organisms. They act as scaffold molecules that regulate the traffic and function of other membrane/signaling proteins, resulting in important downstream cellular consequences. The aim of this work was to use transcriptomes and bioinformatics analysis to identify the tetraspanins (and their partners) involved in trophoblast differentiation. We built a protein-protein interaction network around tetraspanins which revealed that tetraspanins CD9, CD81, and CD82 show a specific expression during trophoblast differentiation. These proteins appeared to be interconnected and to recruit several membrane partners which include integrins, immune-related molecules, and a variety of receptors. During weeks 8 to 24, a CD9 expression trajectory was identified in extravillous trophoblasts, and a website was developed: ( https://extravillous.shinyapps.io/CD9humanEVT/ ). In conclusion, CD81 may, together with CD9 and CD82, be interconnected in controlling trophoblast invasion in the endometrium. CD9 expression trajectory in extravillous trophoblast between weeks 8 and 24 shows the involvement of CD9 in cell adhesion and migration.
Collapse
Affiliation(s)
- Christophe Desterke
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France.,INSERM UA9 Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Villejuif, France
| | - Julien Dang
- INSERM U970, 56 rue Leblanc, 75015, Paris, France.,Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicetre, France
| | - Hans-Kristian Lorenzo
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France.,Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicetre, France.,INSERM U1197, Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Bâtiment Lavoisier, France
| | - Jean-Jacques Candelier
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France. .,INSERM U1197, Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Bâtiment Lavoisier, France.
| |
Collapse
|
46
|
Mittal R, Bencie N, Langlie J, Mittal J, Eshraghi AA. Exosomes as drug delivery vehicles and biomarkers for neurological and auditory systems. J Cell Physiol 2021; 236:8035-8049. [PMID: 34224589 DOI: 10.1002/jcp.30484] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
Exosomes are small extracellular membrane particles that play a crucial role in intracellular signaling. Research shows that exosomes have the potential to be used as biomarkers or drug delivery systems in specific organs, such as the neurological system and the inner ear. Exosomes in neurological and auditory systems release different molecules when under stress versus in healthy states, highlighting their potential use as biomarkers in the identification of diseased states. Studies have suggested that exosomes can be harnessed for drug delivery to hard-to-reach organs, such as cochlear sensory hair cells and the brain due to their ability to cross the blood-labyrinth and blood-brain barriers. In this article, we describe the biogenesis, classification, and characterization methods of exosomes. We then discuss recent studies that indicate their potential usage as biomarkers and drug delivery systems to help treat inner ear and neurological disorders.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nicole Bencie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jake Langlie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Adrien A Eshraghi
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
47
|
Bordanaba-Florit G, Royo F, Kruglik SG, Falcón-Pérez JM. Using single-vesicle technologies to unravel the heterogeneity of extracellular vesicles. Nat Protoc 2021; 16:3163-3185. [PMID: 34135505 DOI: 10.1038/s41596-021-00551-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are heterogeneous lipid containers with a complex molecular cargo comprising several populations with unique roles in biological processes. These vesicles are closely associated with specific physiological features, which makes them invaluable in the detection and monitoring of various diseases. EVs play a key role in pathophysiological processes by actively triggering genetic or metabolic responses. However, the heterogeneity of their structure and composition hinders their application in medical diagnosis and therapies. This diversity makes it difficult to establish their exact physiological roles, and the functions and composition of different EV (sub)populations. Ensemble averaging approaches currently employed for EV characterization, such as western blotting or 'omics' technologies, tend to obscure rather than reveal these heterogeneities. Recent developments in single-vesicle analysis have made it possible to overcome these limitations and have facilitated the development of practical clinical applications. In this review, we discuss the benefits and challenges inherent to the current methods for the analysis of single vesicles and review the contribution of these approaches to the understanding of EV biology. We describe the contributions of these recent technological advances to the characterization and phenotyping of EVs, examination of the role of EVs in cell-to-cell communication pathways and the identification and validation of EVs as disease biomarkers. Finally, we discuss the potential of innovative single-vesicle imaging and analysis methodologies using microfluidic devices, which promise to deliver rapid and effective basic and practical applications for minimally invasive prognosis systems.
Collapse
Affiliation(s)
- Guillermo Bordanaba-Florit
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.
| | - Félix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Madrid, Spain
| | - Sergei G Kruglik
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin, Paris, France
| | - Juan M Falcón-Pérez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Madrid, Spain. .,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
48
|
Awoyemi T, Motta-Mejia C, Zhang W, Kouser L, White K, Kandzija N, Alhamlan FS, Cribbs AP, Tannetta D, Mazey E, Redman C, Kishore U, Vatish M. Syncytiotrophoblast Extracellular Vesicles From Late-Onset Preeclampsia Placentae Suppress Pro-Inflammatory Immune Response in THP-1 Macrophages. Front Immunol 2021; 12:676056. [PMID: 34163477 PMCID: PMC8215361 DOI: 10.3389/fimmu.2021.676056] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/28/2021] [Indexed: 01/08/2023] Open
Abstract
Syncytiotrophoblast derived Extracellular Vesicles (STBEV) from normal pregnancy (NP) have previously been shown to interact with circulating monocytes and B cells and induce pro-inflammatory cytokine release. Early-onset preeclampsia (EOPE) is associated with an exacerbated inflammatory response, yet there is little data regarding late-onset PE (LOPE) and immune function. Here, using a macrophage/monocyte cell line THP-1, we investigated the inflammatory potential of STBEV, comprising medium/large-STBEV (>200nm) and small-STBEV (<200nm), isolated from LOPE (n=6) and normal (NP) (n=6) placentae via dual-lobe ex-vivo placental perfusion and differential centrifugation. THP-1 cells bound and internalised STBEV isolated from NP and LOPE placentae, as revealed by flow cytometry, confocal microscopy, and ELISA. STBEV-treated THP-1 cells were examined for cytokine gene expression by RT-qPCR and the cell culture media examined for secreted cytokines/chemokines. As expected, NP medium/large-STBEV significantly upregulated the transcriptional expression of TNF-α, IL-10, IL-6, IL-12, IL-8 and TGF-β compared to PE medium/large-STBEV. However, there was no significant difference in the small STBEV population between the two groups, although in general, NP small STBEVs slightly upregulated the same cytokines. In contrast, LOPE STBEV (medium and large) did not induce pro-inflammatory responses by differentiated THP-1 macrophages. This decreased effect of LOPE STBEV was echoed in cytokine/chemokine release. Our results appear to suggest that STBEV from LOPE placentae do not have a major immune-modulatory effect on macrophages. In contrast, NP STBEV caused THP-1 cells to release pro-inflammatory cytokines. Thus, syncytiotrophoblast extracellular vesicles from LOPE dampen immune functions of THP-1 macrophages, suggesting an alternative mechanism leading to the pro-inflammatory environment observed in LOPE.
Collapse
Affiliation(s)
- Toluwalase Awoyemi
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Carolina Motta-Mejia
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.,Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Wei Zhang
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Lubna Kouser
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kirsten White
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Neva Kandzija
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Adam P Cribbs
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Dionne Tannetta
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Emily Mazey
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Christopher Redman
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Pregnancy-Related Extracellular Vesicles Revisited. Int J Mol Sci 2021; 22:ijms22083904. [PMID: 33918880 PMCID: PMC8068855 DOI: 10.3390/ijms22083904] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/20/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are small vesicles ranging from 20–200 nm to 10 μm in diameter that are discharged and taken in by many different types of cells. Depending on the nature and quantity of their content—which generally includes proteins, lipids as well as microRNAs (miRNAs), messenger-RNA (mRNA), and DNA—these particles can bring about functional modifications in the receiving cells. During pregnancy, placenta and/or fetal-derived EVs have recently been isolated, eliciting interest in discovering their clinical significance. To date, various studies have associated variations in the circulating levels of maternal and fetal EVs and their contents, with complications including gestational diabetes and preeclampsia, ultimately leading to adverse pregnancy outcomes. Furthermore, EVs have also been identified as messengers and important players in viral infections during pregnancy, as well as in various congenital malformations. Their presence can be detected in the maternal blood from the first trimester and their level increases towards term, thus acting as liquid biopsies that give invaluable insight into the status of the feto-placental unit. However, their exact roles in the metabolic and vascular adaptations associated with physiological and pathological pregnancy is still under investigation. Analyzing peer-reviewed journal articles available in online databases, the purpose of this review is to synthesize current knowledge regarding the utility of quantification of pregnancy related EVs in general and placental EVs in particular as non-invasive evidence of placental dysfunction and adverse pregnancy outcomes, and to develop the current understanding of these particles and their applicability in clinical practice.
Collapse
|
50
|
Syncytiotrophoblast Derived Extracellular Vesicles in Relation to Preeclampsia. MATERNAL-FETAL MEDICINE 2021. [DOI: 10.1097/fm9.0000000000000093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|