1
|
Mattei DN, Harman RM, Van de Walle GR, Smith R, Grivel JC, Abdelalim EM, Vinardell T. Effect of pregnancy on isolation efficiency and in vitro proliferation of equine peripheral-blood derived mesenchymal stromal cells. Theriogenology 2024; 224:107-118. [PMID: 38761667 DOI: 10.1016/j.theriogenology.2024.05.015] [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: 01/29/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Mesenchymal stromal cells (MSCs) have regenerative and immunomodulatory potential and may be used to treat injured tissues. Pregnancy has been associated with increased MSCs in the peripheral circulation in multiple species, but to date, there are no reports on this matter in horses. This study aimed to evaluate the effect of pregnancy on isolation efficiency and proliferation capacity of equine MSCs derived from the peripheral blood (PB) of mares. Venous blood samples were collected at the 11th month of gestation and 1 month after delivery from clinically healthy Arabian mares that presented normal pregnancies. Blood samples were processed for in vitro cellular culture and hormonal and metabolic profiles. MSCs were isolated and characterized by trilineage differentiation potential, immunophenotyping, analyzed by gene sequencing and proliferation assays. The isolation of peripheral blood mononuclear cells (PBMCs) of pregnant mares were associated with higher isolation efficiency and proliferative capacity of MSCs derived from peripheral blood (PB-MSCs) recovered pre-partum than those isolated post-partum. Although fetal gender, parity, 5α-reduced pregnanes, insulin, and cortisol were shown to affect cellular proliferation, individual factors and the small population studied must be considered. This study suggests that PB-MSCs from pregnant mares could be a valuable alternative source of MSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Debora N Mattei
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar
| | - Rebecca M Harman
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Gerlinde R Van de Walle
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Roger Smith
- Department of Clinical Science and Services, The Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, United Kingdom
| | - Jean Charles Grivel
- Deep Phenotyping Core, Sidra Medicine, PO Box 26999, Al Garrafa St, Ar-Rayyan, Doha, Qatar
| | - Essam M Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Division, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar; Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar
| | - Tatiana Vinardell
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar.
| |
Collapse
|
2
|
Kontovazainitis CG, Gialamprinou D, Theodoridis T, Mitsiakos G. Hemostasis in Pre-Eclamptic Women and Their Offspring: Current Knowledge and Hemostasis Assessment with Viscoelastic Tests. Diagnostics (Basel) 2024; 14:347. [PMID: 38337863 PMCID: PMC10855316 DOI: 10.3390/diagnostics14030347] [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: 12/15/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Pre-eclampsia (PE) is a placenta-mediated disease and remains a major cause of maternal and neonatal mortality and morbidity. As PE develops, normal pregnancy's hypercoagulable balance is disrupted, leading to platelet hyperactivation, excessive pathological hypercoagulability, and perturbed fibrinolysis. This narrative review aims to summarize the current knowledge regarding hemostasis in PE compared with healthy gestation and the potential effects of maternal PE on neonatal hemostasis. Finally, it aims to discuss hemostasis assessments for normal pregnancies and PE, emphasizing the role of viscoelastic tests, namely, thromboelastography (TEG) and thromboelastometry (ROTEM), for monitoring PE-associated hemostatic alterations. The use of TEG/ROTEM for assessing the hemostatic profile of PE women has been little considered, even though conventional coagulation tests (CCTs) have not helped to monitor hemostasis in this population. Compared with normal pregnancy, TEG/ROTEM in PE reveals an excessive hypercoagulability analogous with the severity of the disease, characterized by higher-stability fibrin clots. The TEG/ROTEM parameters can reflect PE severity and may be used for monitoring and as predictive markers for the disease.
Collapse
Affiliation(s)
- Christos-Georgios Kontovazainitis
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| | - Dimitra Gialamprinou
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| | - Theodoros Theodoridis
- 1st Department of Obstetrics and Gynecology, “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece;
| | - Georgios Mitsiakos
- 2nd Neonatal Department and Neonatal Intensive Care Unit (NICU), “Papageorgiou” University Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece; (C.-G.K.); (D.G.)
| |
Collapse
|
3
|
Chen Y, Wan G, Li Z, Liu X, Zhao Y, Zou L, Liu W. Endothelial progenitor cells in pregnancy-related diseases. Clin Sci (Lond) 2023; 137:1699-1719. [PMID: 37986615 PMCID: PMC10665129 DOI: 10.1042/cs20230853] [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: 08/01/2023] [Revised: 10/09/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Placental neovascularization plays a crucial role in fetomaternal circulation throughout pregnancy and is dysregulated in several pregnancy-related diseases, including preeclampsia, gestational diabetes mellitus, and fetal growth restriction. Endothelial progenitor cells (EPCs) are a heterogeneous population of cells that differentiate into mature endothelial cells, which influence vascular homeostasis, neovascularization, and endothelial repair. Since their discovery in 1997 by Asahara et al., the role of EPCs in vascular biology has garnered a lot of interest. However, although pregnancy-related conditions are associated with changes in the number and function of EPCs, the reported findings are conflicting. This review discusses the discovery, isolation, and classification of EPCs and highlights discrepancies between current studies. Overviews of how various diseases affect the numbers and functions of EPCs, the role of EPCs as biomarkers of pregnancy disorders, and the potential therapeutic applications involving EPCs are also provided.
Collapse
Affiliation(s)
- Yangyang Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zeyun Li
- The First Clinical School of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yin Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weifang Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
4
|
Giri J, Modi D. Endometrial and placental stem cells in successful and pathological pregnancies. J Assist Reprod Genet 2023; 40:1509-1522. [PMID: 37338750 PMCID: PMC10352206 DOI: 10.1007/s10815-023-02856-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
The endometrium is a dynamic tissue that undergoes extensive remodeling during the menstrual cycle and further gets modified during pregnancy. Different kinds of stem cells are reported in the endometrium. These include epithelial stem cells, endometrial mesenchymal stem cells, side population stem cells, and very small embryonic-like stem cells. Stem cells are also reported in the placenta which includes trophoblast stem cells, side population trophoblast stem cells, and placental mesenchymal stem cells. The endometrial and placental stem cells play a pivotal role in endometrial remodeling and placental vasculogenesis during pregnancy. The dysregulation of stem cell function is reported in various pregnancy complications like preeclampsia, fetal growth restriction, and preterm birth. However, the mechanisms by which it does so are yet elusive. Herein, we review the current knowledge of the different type of stem cells involved in pregnancy initiation and also highlight how their improper functionality leads to pathological pregnancy.
Collapse
Affiliation(s)
- Jayeeta Giri
- Molecular and Cellular Biology Laboratory, ICMR-National Institute for Research in Reproductive and child Health, Indian Council of Medical Research (ICMR), JM Street, Parel, Mumbai, 400012, India.
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, ICMR-National Institute for Research in Reproductive and child Health, Indian Council of Medical Research (ICMR), JM Street, Parel, Mumbai, 400012, India.
| |
Collapse
|
5
|
Singh A, Jaiswar SP, Priyadarshini A, Deo S. Reduced Endothelial Progenitor Cells: A Possible Biomarker for Idiopathic Fetal Growth Restriction in Human Pregnancies. JOURNAL OF MOTHER AND CHILD 2023; 27:182-189. [PMID: 37991978 PMCID: PMC10664836 DOI: 10.34763/jmotherandchild.20232701.d-23-00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 10/06/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Circulating endothelial progenitor cells (EPCs) may be necessary throughout pregnancy by ensuring proper placentation and embryonic growth. The lack of standardized EPC quantification techniques has prevented conclusive proof of an increase in EPC during pregnancy. OBJECTIVES The purpose of this study was to determine whether EPC levels change for healthy and idiopathic fetal growth restriction (FGR) pregnancies. MATERIALS AND METHODS The study population consisted of 48 healthy pregnant females with no previous history of IUGR (10 in the first trimester, 15 in the second, and 23 in the third), 48 women with pregnancy complicated by idiopathic FGR, and 15 non-pregnant women. By using flow cytometry, EPCs in maternal blood were recognized as CD45dim/CD34/KDR cells. ELISA was used to measure plasmatic cytokines. RESULTS We ascertained a progressive rise in EPCs in healthy pregnancies that was apparent in the first but more pronounced in the third trimester. At comparable gestational ages, FGR-complicated pregnancies had impaired EPC growth. Placental growth factor and stromal-derived factor-1 levels in the blood were significantly lower in FGR than in healthy pregnancies, which may have contributed to the degradation of the EPCs. CONCLUSION The count in EPCs might hold considerable promise toward developing a peculiar authentication marker for observing pregnancies, and could be the focus of cutting-edge tactics for the prognosis and treatment of FGR pregnancies.
Collapse
Affiliation(s)
- Apurva Singh
- Department of Obstetrics and Gynaecology, King George's Medical University, Lucknow
- Photobiology Division, CSIR-Indian Institute of Toxicology Research, Lucknow
| | - Shyam Pyari Jaiswar
- Department of Obstetrics and Gynaecology, King George's Medical University, Lucknow
| | - Apala Priyadarshini
- Department of Obstetrics and Gynaecology, King George's Medical University, Lucknow
| | - Sujata Deo
- Department of Obstetrics and Gynaecology, King George's Medical University, Lucknow
| |
Collapse
|
6
|
McCartney SA, Kolarova T, Kanaan SB, Chae A, Laughney CI, Nelson JL, Gammill HS, Shree R. Increased fetal microchimerism in immune and stem cell subsets in preeclampsia. Am J Reprod Immunol 2023; 89:e13666. [PMID: 36482289 PMCID: PMC10413445 DOI: 10.1111/aji.13666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
PROBLEM Preeclampsia (PE) is associated with an increased risk of maternal cardiovascular disease (CVD), however, it is unclear whether this is due to shared underlying physiology or changes which occur during the disease process. Fetal microchimerism (FMc) within the maternal circulation can durably persist decades after pregnancy, is known to occur at greater frequency in PE, and can potentially affect local and systemic immune programming, thus changes in cellular FMc may provide a mechanism for long-term health outcomes associated with PE. METHOD OF STUDY We investigated whether PE is associated with alterations in FMc immune and stem cell populations. We analyzed maternal peripheral blood mononuclear cells (PBMC) from PE cases (n = 16) and matched controls from normal pregnancies (n = 16), from which immune and stem cell subsets were isolated by flow cytometry. Genomic DNA was extracted from total PMBC and individual cell subsets, and FMc frequency was quantified by quantitative polymerase chain reaction assays targeting a fetal-specific non-shared polymorphism identified from family genotyping. RESULTS There was a significant increase in FMc concentration in immune cell subsets in PE cases compared to controls, predominantly in B cell, and NK cell lymphocyte populations. There was no significant difference in FMc frequency or concentration within the stem cell population between PE and controls. CONCLUSIONS The altered concentrations of immune cells within FMc in the maternal blood provides a potential mechanism for the inflammation which occurs during PE to induce long-lasting changes to the maternal immune system and may potentially promote chronic maternal disease.
Collapse
Affiliation(s)
- Stephen A McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Teodora Kolarova
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Sami B Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Angel Chae
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Caitlin I Laughney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - J Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hilary S Gammill
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Raj Shree
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Lin A, Pehrson M, Sarno G, Fraser A, Rich-Edwards JW, Gonҫalves I, Pihlsgård M, Timpka S. Coronary Artery Restenosis in Women by History of Preeclampsia. J Am Heart Assoc 2022; 11:e026287. [PMID: 36073639 DOI: 10.1161/jaha.122.026287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background A history of preeclampsia is associated with increased risk of coronary artery disease and experimental evidence suggests that a history of preeclampsia also increases the risk of restenosis. However, the extent to which a history of preeclampsia is associated with risk of restenosis after percutaneous coronary intervention in women is unknown. Methods and Results We included 6065 parous women aged ≤65 years with first percutaneous coronary intervention on 9452 segments 2006 to 2017, linking nationwide data on percutaneous coronary intervention and delivery history in Sweden. Main outcomes were clinical restenosis and target lesion revascularization within 2 years. We accounted for segment-, procedure-, and patient-related potential predictors of restenosis in proportional hazards regression models. Restenosis occurred in 345 segments (3.7%) and target lesion revascularization was performed on 383 patients (6.3%). A history of preeclampsia was neither significantly associated with risk of restenosis (predictor-accounted hazard ratio [HR], 0.71 [95% CI, 0.41-1.23]) nor target lesion revascularization (0.74 [95% CI, 0.51-1.07]) compared with a normotensive pregnancy history. When term and preterm preeclampsia were investigated separately, segments in women with a history of term preeclampsia had a lower risk of restenosis (predictor-accounted HR, 0.45 [95% CI, 0.21-0.94]). A history of preeclampsia was not significantly associated with death by any cause within 2 years of the index procedure (predictor-accounted HR 1.06, [95% CI, 0.62-1.80]). Conclusions A history of preeclampsia was not associated with increased risk of restenosis but instead some evidence pointed to a decreased risk. To facilitate future studies and allow for replication, concomitant collection of data on pregnancy complication history and percutaneous coronary intervention outcomes in women is warranted.
Collapse
Affiliation(s)
- Annie Lin
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Moa Pehrson
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Giovanna Sarno
- Department of Medical Sciences Cardiology and Uppsala Clinical Research Center, Uppsala University Uppsala Sweden
| | - Abigail Fraser
- Population Health Science, Bristol Medical School University of Bristol Bristol United Kingdom
| | - Janet W Rich-Edwards
- Division of Women's Health Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA
| | - Isabel Gonҫalves
- Department of Cardiology and Cardiovascular Research Translational Studies Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Mats Pihlsgård
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Simon Timpka
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden.,Department of Obstetrics and Gynecology Skåne University Hospital Malmö Sweden
| |
Collapse
|
8
|
Simoncini S, Coppola H, Rocca A, Bachmann I, Guillot E, Zippo L, Dignat-George F, Sabatier F, Bedel R, Wilson A, Rosenblatt-Velin N, Armengaud JB, Menétrey S, Peyter AC, Simeoni U, Yzydorczyk C. Endothelial Colony-Forming Cells Dysfunctions Are Associated with Arterial Hypertension in a Rat Model of Intrauterine Growth Restriction. Int J Mol Sci 2021; 22:10159. [PMID: 34576323 PMCID: PMC8465555 DOI: 10.3390/ijms221810159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
Infants born after intrauterine growth restriction (IUGR) are at risk of developing arterial hypertension at adulthood. The endothelium plays a major role in the pathogenesis of hypertension. Endothelial colony-forming cells (ECFCs), critical circulating components of the endothelium, are involved in vasculo-and angiogenesis and in endothelium repair. We previously described impaired functionality of ECFCs in cord blood of low-birth-weight newborns. However, whether early ECFC alterations persist thereafter and could be associated with hypertension in individuals born after IUGR remains unknown. A rat model of IUGR was induced by a maternal low-protein diet during gestation versus a control (CTRL) diet. In six-month-old offspring, only IUGR males have increased systolic blood pressure (tail-cuff plethysmography) and microvascular rarefaction (immunofluorescence). ECFCs isolated from bone marrow of IUGR versus CTRL males displayed a decreased proportion of CD31+ versus CD146+ staining on CD45- cells, CD34 expression (flow cytometry, immunofluorescence), reduced proliferation (BrdU incorporation), and an impaired capacity to form capillary-like structures (Matrigel test), associated with an impaired angiogenic profile (immunofluorescence). These dysfunctions were associated with oxidative stress (increased superoxide anion levels (fluorescent dye), decreased superoxide dismutase protein expression, increased DNA damage (immunofluorescence), and stress-induced premature senescence (SIPS; increased beta-galactosidase activity, increased p16INK4a, and decreased sirtuin-1 protein expression). This study demonstrated an impaired functionality of ECFCs at adulthood associated with arterial hypertension in individuals born after IUGR.
Collapse
Affiliation(s)
- Stephanie Simoncini
- Aix Marseille Univ, Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAe), Center from Cardiovascular and Nutrition research (C2VN), UMR-S 1263, UFR de Pharmacie, Campus Santé, 13385 Marseille, France; (S.S.); (F.D.-G.); (F.S.)
| | - Hanna Coppola
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Angela Rocca
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Isaline Bachmann
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Estelle Guillot
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Leila Zippo
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Françoise Dignat-George
- Aix Marseille Univ, Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAe), Center from Cardiovascular and Nutrition research (C2VN), UMR-S 1263, UFR de Pharmacie, Campus Santé, 13385 Marseille, France; (S.S.); (F.D.-G.); (F.S.)
| | - Florence Sabatier
- Aix Marseille Univ, Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAe), Center from Cardiovascular and Nutrition research (C2VN), UMR-S 1263, UFR de Pharmacie, Campus Santé, 13385 Marseille, France; (S.S.); (F.D.-G.); (F.S.)
| | - Romain Bedel
- Flow Cytometry Facility, Department of Formation and Research, University of Lausanne, 1011 Lausanne, Switzerland; (R.B.); (A.W.)
| | - Anne Wilson
- Flow Cytometry Facility, Department of Formation and Research, University of Lausanne, 1011 Lausanne, Switzerland; (R.B.); (A.W.)
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland
| | - Nathalie Rosenblatt-Velin
- Department Heart-Vessels, Division of Angiology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland;
| | - Jean-Baptiste Armengaud
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Steeve Menétrey
- Department Woman-Mother-Child, Neonatal Research Laboratory, Clinic of Neonatology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.M.); (A.-C.P.)
| | - Anne-Christine Peyter
- Department Woman-Mother-Child, Neonatal Research Laboratory, Clinic of Neonatology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.M.); (A.-C.P.)
| | - Umberto Simeoni
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| | - Catherine Yzydorczyk
- Department Woman-Mother-Child, Division of pediatrics, DOHaD Laboratory, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (H.C.); (A.R.); (I.B.); (E.G.); (L.Z.); (J.-B.A.); (U.S.)
| |
Collapse
|
9
|
Brodowski L, Schröder-Heurich B, von Hardenberg S, Richter K, von Kaisenberg CS, Dittrich-Breiholz O, Meyer N, Dörk T, von Versen-Höynck F. MicroRNA Profiles of Maternal and Neonatal Endothelial Progenitor Cells in Preeclampsia. Int J Mol Sci 2021; 22:ijms22105320. [PMID: 34070163 PMCID: PMC8158476 DOI: 10.3390/ijms22105320] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/08/2023] Open
Abstract
Preeclampsia is associated with an increased cardiovascular morbidity of mother and offspring, thus contributing to a substantial burden in women and children’s health. It has been proven that endothelial progenitor cell (EPC) numbers and functional characteristics are impaired in cardiovascular disease and preeclampsia, although causative factors for the latter have remained elusive. MicroRNA (miRNA) modifications are a potential mechanism through which exposure to an altered environment translates into the development of chronic disease. In this study, we examined whether development of preeclampsia corresponds to alterations of miRNAs in maternal- and cord-blood-derived EPC. To test this end, we analyzed maternal and neonatal miRNAs via RNA sequencing from endothelial cells of preeclamptic and healthy controls in different cell culture passages. We were able to demonstrate differentially represented miRNAs in all groups. Hsa-miR-1270 showed significantly different levels in cord blood EPC from preeclampsia versus control and was negatively correlated with mRNA levels of its predicted targets ANGPTL7 and TFRC. Transfection with an hsa-miR-1270 inhibitor decreased the tube formation capacity and chemotactic motility but did not change proliferation in vitro. Target predictions and gene set enrichment analyses identified alternative splicing as a significantly enriched pathway for hsa-miR-1270. The top miRNAs in three other groups were predicted to target transcriptional and developmental pathways. Here, we showed for the first time significantly different levels of miRNAs and differently represented mRNA levels of predicted target genes in EPC derived from preeclampsia. Understanding the effects of preeclampsia on the epigenetic mechanisms of EPC will be crucial and may provide initial insights for further evaluation of the benefits of therapies targeting this cell population.
Collapse
Affiliation(s)
- Lars Brodowski
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| | - Bianca Schröder-Heurich
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
| | - Sandra von Hardenberg
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
| | - Katja Richter
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
| | - Constantin S. von Kaisenberg
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| | - Oliver Dittrich-Breiholz
- Research Core Unit Genomics, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| | - Nadia Meyer
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
| | - Thilo Dörk
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
| | - Frauke von Versen-Höynck
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (L.B.); (B.S.-H.); (S.v.H.); (K.R.); (N.M.); (T.D.)
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
- Correspondence: ; Tel.: +49-511-532-8703; Fax: +49-511-532-6081
| |
Collapse
|
10
|
Neurology of Preeclampsia and Related Disorders: an Update in Neuro-obstetrics. Curr Pain Headache Rep 2021; 25:40. [PMID: 33825997 PMCID: PMC10069269 DOI: 10.1007/s11916-021-00958-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Preeclampsia and related hypertensive disorders of pregnancy affect up to 10% of pregnancies. Neurological complications are common and neurologists often become involved in the care of obstetric patients with preeclampsia. Here, we review the definition(s), epidemiology, clinical features, and pathophysiology of preeclampsia, focusing on maternal neurological complications and headache as a common presenting symptom of preeclampsia. RECENT FINDINGS Neurological symptoms are early and disease-defining features of preeclampsia. Neurological complications of preeclampsia may include headaches, visual symptoms, cerebral edema, seizures, or acute cerebrovascular disorders such as intracerebral hemorrhage or reversible cerebral vasoconstriction syndrome. A history of migraine is an independent risk factor for vascular diseases during pregnancy, including preeclampsia and maternal stroke. The pathophysiology of both preeclampsia and migraine is complex, and the mechanisms linking the two are not fully understood. Overlapping clinical and pathophysiological features of migraine and preeclampsia include inflammation, vascular endothelial dysfunction, and changes in vasoreactivity. Neurological complications are recognized as a major contributor to maternal morbidity and mortality. Pregnant and postpartum women commonly present with headache, and red flags in the clinical history and examination should prompt urgent neuroimaging and laboratory evaluation. A focused headache history should be elicited from patients as part of routine obstetrical care to identify patients at an increased risk of preeclampsia and related hypertensive disorders of pregnancy. Collaborative models of care and scientific investigation in the emerging field of neuro-obstetrics have the common goal of reducing the risk of maternal neurological morbidity and mortality from preeclampsia.
Collapse
|
11
|
Reduced angiovasculogenic and increased inflammatory profiles of cord blood cells in severe but not mild preeclampsia. Sci Rep 2021; 11:3630. [PMID: 33574435 PMCID: PMC7878804 DOI: 10.1038/s41598-021-83146-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 01/22/2021] [Indexed: 11/17/2022] Open
Abstract
Preeclampsia (PE) is a prevalent pregnancy disorder that leads to high maternal and fetal morbidity and mortality. While defective vascular development and angiogenesis in placenta are known as crucial pathological findings, its pathophysiological mechanism remains elusive. To better understand the effects of PE on angio-vasculogenesis and inflammatory networks in the fetus and to identify their biological signatures, we investigated the quantitative and functional characteristics of cord blood-derived mononuclear cells (CB-MNCs) and CD31-positive MNCs. Flow cytometry analysis demonstrated that the CB-MNCs from the severe PE group had significantly decreased number of cells expressing CD3, CD11b, CD14, CD19, KDR, and CD31 compared with the normal group. Quantitative real time PCR (qRT-PCR) shows down-regulation of the major angiogenic factor VEGFA in MNCs and CD31+ MNCs in severe PE. The major inflammatory cytokines IL1 was highly upregulated in CD31+ CB-MNCs in the severe PE patients. Mild PE patients, however, did not display any significant difference in expression of all measured angiogenic genes and most inflammatory genes. These findings show distinct angiogenic and inflammatory signatures from severe PE, and they may play a significant role in the pathogenesis of vascular defects in placenta of severe PE.
Collapse
|
12
|
Mallucci G, Beneventi F, Bergamaschi R, Bizzotto C, Cavagnoli C, De Maggio I, Bellingeri C, Monti C, Viarengo G, Spinillo A. Circulating endothelial progenitor cells during pregnancy in multiple sclerosis. Neurol Sci 2020; 42:1443-1451. [PMID: 32804349 PMCID: PMC7956006 DOI: 10.1007/s10072-020-04648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 08/01/2020] [Indexed: 11/30/2022]
Abstract
Background Endothelial progenitor cells (EPCs) have been shown to increase during physiological pregnancy and are believed to play a fundamental role in the process of placentation. Reduced levels of EPCs during pregnancy have been associated with preeclampsia and miscarriage. Women with multiple sclerosis (MS) are not at increased risk of preeclampsia nor of general adverse obstetric outcome, in contrast with some other autoimmune diseases. Objective The aim of this study was to evaluate circulating EPCs levels in pregnant patients with MS. Methods CD34+ and CD133+ were longitudinally detected by flow cytometry in the maternal plasma of 29 healthy controls and 9 MS patients and in the cord blood of their newborns. Results EPCs were affected by pregnancy with the same trend in both groups (CD34+ p = 0.0342; CD133+ p = 0.0347). EPCs during pregnancy were increased in MS (mean ± SD: CD34+ cells 0.038 ± 0.010; CD133+ 0.024 ± 0.009) with respect to healthy controls (mean ± SD: CD34+ cells 0.022 ± 0.006; CD133+ 0.016 ± 0.004), CD34+ p = 0.0004; CD133+ p = 0.0109. EPCs levels of the cord blood of MS patients' newborns mild correlated with maternal EPC levels at delivery (CD34+: spearman’s Rho 0.658, p = 0.054; CD133+: spearman’s Rho 0.758, p = 0.018). Conclusions This work identified increased circulating EPC levels during pregnancy, following the same trend both in MS patients and healthy controls. Despite the similar trend, the levels of circulating EPCs were significantly higher in MS patients with respect to the control population. A correlation was also found in MS patients between cord blood EPCs and circulating EPCs at delivery. Electronic supplementary material The online version of this article (10.1007/s10072-020-04648-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giulia Mallucci
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
- Multiple Sclerosis Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy.
| | - Fausta Beneventi
- Department of Obstetrics and Gynaecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Roberto Bergamaschi
- Multiple Sclerosis Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
| | - Cristina Bizzotto
- Multiple Sclerosis Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
| | - Chiara Cavagnoli
- Department of Obstetrics and Gynaecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Irene De Maggio
- Department of Obstetrics and Gynaecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Camilla Bellingeri
- Department of Obstetrics and Gynaecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Cristina Monti
- Department of Public Health Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Gianluca Viarengo
- Immunohaematology and Transfusion Service, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynaecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
13
|
Kim JH, Kim JY, Park M, Kim S, Kim T, Kim J, Choi S, Park W, Hwang JY, Choe J, Ha KS, Won MH, Ryoo S, Kwon YG, Kim YM. NF-κB-dependent miR-31/155 biogenesis is essential for TNF-α-induced impairment of endothelial progenitor cell function. Exp Mol Med 2020; 52:1298-1309. [PMID: 32770080 PMCID: PMC8080610 DOI: 10.1038/s12276-020-0478-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/16/2020] [Accepted: 04/01/2020] [Indexed: 01/31/2023] Open
Abstract
Endothelial progenitor cell (EPC) dysfunction impairs vascular function and remodeling in inflammation-associated diseases, including preeclampsia. However, the underlying mechanism of this inflammation-induced dysfunction remains unclear. In the present study, we found increases in TNF-α and miR-31/155 levels and reduced numbers of circulating EPCs in patients with preeclampsia. Patient-derived mononuclear cells (MNCs) cultured in autologous serum had decreased endothelial nitric oxide synthase (eNOS) expression, nitric oxide production, and differentiation into EPCs with angiogenic potential, and these effects were inhibited by a TNF-α-neutralizing antibody and miR-31/155 inhibitors. Moreover, TNF-α treatment of normal MNCs increased miR-31/155 biogenesis, decreased eNOS expression, reduced EPC differentiation, and impaired angiogenic potential. The TNF-α-induced impairment of EPC differentiation and function was rescued by NF-κB p65 knockdown or miR-31/155 inhibitors. In addition, treatment of MNCs with synthetic miR-31/155 or an eNOS inhibitor mimicked the inhibitory effects of TNF-α on eNOS expression and EPC functions. Moreover, transplantation of EPCs that had been differentiated from TNF-α-treated MNCs decreased neovascularization and blood perfusion in ischemic mouse hindlimbs compared with those of normally differentiated EPCs. These findings suggest that NF-κB activation is required for TNF-α-induced impairment of EPC mobilization, differentiation, and function via miR-31/155 biogenesis and eNOS downregulation. Our data provide a new role for NF-κB-dependent miR-31/155 in EPC dysfunction under the pathogenic conditions of inflammation-associated vascular diseases, including preeclampsia. miRNA molecules that inhibit the activity of specific genes are implicated in a cellular control network involved in some of the damaging effects of inflammation on blood vessels. Researchers in South Korea led by Young-Myeong Kim at Kangwon National University School of Medicine, Chuncheon, identified the link by studying cells from patients with the inflammatory condition pre-eclampsia, characterized by hypertension. They found that two miRNAs, miR-31 and miR-155, are involved in molecular signaling processes that impair the production of endothelial progenitor cells lining blood vessels, which is essential for maintenance and repair. The research also identified the key protein eNOS involved in the miRNA molecules’ mechanism of action. Understanding these miRNAs and the protein involved in their production and action may help researchers develop new treatments for blood vessel diseases associated with inflammation.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Minsik Park
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Suji Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Taesam Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Joohwan Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Seunghwan Choi
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Wonjin Park
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Jong Yun Hwang
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Jongseon Choe
- Department of Immunology, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea
| | - Sungwoo Ryoo
- Department of Life Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea.
| |
Collapse
|
14
|
High mobilization of CD133+/CD34+ cells expressing HIF-1α and SDF-1α in septic abdominal surgical patients. BMC Anesthesiol 2020; 20:158. [PMID: 32593288 PMCID: PMC7320250 DOI: 10.1186/s12871-020-01068-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/09/2020] [Indexed: 12/25/2022] Open
Abstract
Background The control of endothelial progenitor cells (CD133+/CD34+ EPCs) migrating from bone marrow to peripheral blood is not completely understood. Emerging evidence suggests that stromal cell-derived factor-1α (SDF-1α) mediates egression of EPCs from bone marrow, while the hypoxia inducible factor (HIF) transcriptional system regulates SDF-1α expression. Our study aimed to investigate the time course of circulating CD133+/CD34+ EPCs and its correlation with the expression of HIF-1α protein and SDF-1α in postoperative laparoscopic abdominal septic patients. Methods Postoperative patients were divided in control (C group) and septic group (S group) operated immediately after the diagnosis of sepsis/septic shock. Blood samples were collected at baseline (0), 1, 3 and 7 postoperative days for CD133+/CD34+ EPCs count expressing or not the HIF-1α and SDF-1α analysis. Results Thirty-two patients in S group and 39 in C group were analyzed. In C group CD133+/CD34+ EPCs count remained stable throughout the study period, increasing on day 7 (173 [0–421] /μl vs baseline: P = 0.04; vs day 1: P = 0.002). In S group CD133+/CD34+ EPCs count levels were higher on day 3 (vs day 1: P = 0.006 and day 7: P = 0.026). HIF-1α expressing CD133+/CD34+ EPCs count decreased on day 1 as compared with the other days in C group (day 0 vs 1: P = 0.003, days 3 and 7 vs 1: P = 0.008), while it was 321 [0–1418] /μl on day 3 (vs day 1; P = 0.004), and 400 [0–587] /μl on day 7 in S group. SDF-1α levels were higher not only on baseline but also on postoperative day 1 in S vs C group (219 [124–337] pg/ml vs 35 [27–325] pg/ml, respectively; P = 0.01). Conclusion Our results indicate that sepsis in abdominal laparoscopic patients might constitute an additional trigger of the EPCs mobilization as compared with non-septic surgical patients. A larger mobilization of CD133+/CD34+ EPCs, preceded by enhanced plasmatic SDF-1α, occurs in septic surgical patients regardless of HIF-1α expression therein. Trial registration ClinicalTrials.gov no. NCT02589535. Registered 28 October 2015.
Collapse
|
15
|
Wang Y, Lim R, Nie G. Elevated circulating HtrA4 in preeclampsia may alter endothelial expression of senescence genes. Placenta 2019; 90:71-81. [PMID: 32056555 DOI: 10.1016/j.placenta.2019.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Preeclampsia (PE) is a serious complication of human pregnancy. Women who have had PE, especially early-onset PE (EPE), have an increased risk of cardiovascular disease (CVD) later in life. However, how PE is linked to CVD is not well understood. We previously reported that HtrA4, a placenta-specific protease, is significantly elevated in EPE, and inhibits the proliferation of endothelial cells as well as endothelial progenitor cells (EPCs). This can potentially impair endothelial repair and regeneration, leading to endothelial aging, which is a major risk factor of CVD. In this study, we examined whether HtrA4 can alter endothelial expression of senescence genes. METHODS Human umbilical vein endothelial cells (HUVECs) and primary EPCs isolated from cord blood of healthy pregnancies were used as in vitro models. Firstly, HUVECs were treated with HtrA4 at the highest levels detected in EPE for 48h and screened with a senescence PCR array. The results were then validated by RT-PCR and ELISA in HUVECs and EPCs treated with HtrA4 for 24 and 48h. RESULTS We observed that HtrA4 significantly up-regulated IGFBP3, SERPINE1 and SERPINB2, which all promote senescence. IGFBP-3 protein was also significantly elevated in the media of HtrA4-treated HUVECs. Conversely, a number of genes including CDKN2C, PCNA, CALR, CHEK2 and NOX4 were downregulated by HtrA4. Many of these genes also showed a similar trend of change in EPCs following HtrA4 treatment. DISCUSSION Elevation of placenta-derived HtrA4 in PE alters the expression of endothelial genes to promote cellular senescence and may contribute to premature endothelial aging.
Collapse
Affiliation(s)
- Yao Wang
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3168, Australia
| | - Guiying Nie
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
16
|
Brown N, Khan F, Alshaikh B, Berka N, Liacini A, Alawad E, Yusuf K. CD-34 + and VE-cadherin + endothelial progenitor cells in preeclampsia and normotensive pregnancies. Pregnancy Hypertens 2019; 16:42-47. [PMID: 31056159 DOI: 10.1016/j.preghy.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/12/2019] [Accepted: 02/22/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The objective of our study was to determine levels of endothelial progenitor cells (EPCs) in preeclampsia and normotensive pregnant women. STUDY DESIGN Prospective cohort study of women with preeclampsia and normotensive pregnancies. EPCs were estimated by flow cytometry. Multiple linear regression was used to assess the association of EPCs with preeclampsia adjusting for maternal age, body mass index (BMI), gestation and ethnicity. MAIN OUTCOME MEASURE Levels of EPCs in preeclampsia and normotensive pregnancies, with CD-34 and vascular endothelial (VE)-cadherin as markers of EPCs. VE-cadherin is an endothelial cell adhesion molecule used to delineate endothelial lineage of EPCs. RESULTS There were thirty women in the preeclampsia group and thirty-three in the normotensive group. The two groups were similar except for the BMI and blood pressures, which were higher in preeclampsia. On multiple linear regression, EPCs numbers were significantly higher by 29 (95% confidence interval 11.7-46.6, p = 0.001) in preeclampsia compared to the normotensive group. There was significant positive correlation between EPCs and systolic blood pressure in preeclampsia (Spearman correlation coefficient 0.39, p = 0.03). CONCLUSION Although widely used in cardiovascular disease other than preeclampsia, this is the first study using VE-cadherin as a marker of endothelial lineage to define EPCs in preeclampsia. Our results suggest the higher number of EPCs in preeclampsia may be a response of the bone marrow to endothelial injury.
Collapse
Affiliation(s)
- Nicole Brown
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Faisal Khan
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Belal Alshaikh
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Noureddine Berka
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Abdelhamid Liacini
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Essa Alawad
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Kamran Yusuf
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada.
| |
Collapse
|
17
|
Brodowski L, Zindler T, von Hardenberg S, Schröder-Heurich B, von Kaisenberg CS, Frieling H, Hubel CA, Dörk T, von Versen-Höynck F. Preeclampsia-Associated Alteration of DNA Methylation in Fetal Endothelial Progenitor Cells. Front Cell Dev Biol 2019; 7:32. [PMID: 30949477 PMCID: PMC6436196 DOI: 10.3389/fcell.2019.00032] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/25/2019] [Indexed: 01/06/2023] Open
Abstract
Objective The pregnancy complication preeclampsia represents an independent risk factor for cardiovascular disease. Our previous research shows a diminished function of fetal endothelial colony-forming cells (ECFC), a proliferative subgroup of endothelial progenitor cells (EPC) in preeclampsia. The aim of this study was to further investigate whether DNA methylation of fetal EPC is affected in preeclampsia. Methods The genomic methylation pattern of fetal ECFC from uncomplicated and preeclamptic pregnancies was compared for 865918 CpG sites, and genes were classified into gene networks. Low and advanced cell culture passages were compared to explore whether expansion of fetal ECFC in cell culture leads to changes in global methylation status and if methylation characteristics in preeclampsia are maintained with increasing passage. Results A differential methylation pattern of fetal ECFC from preeclampsia compared to uncomplicated pregnancy was detected for a total of 1266 CpG sites in passage 3, and for 2362 sites in passage 5. Key features of primary networks implicated by methylation differences included cell metabolism, cell cycle and transcription and, more specifically, genes involved in cell-cell interaction and Wnt signaling. We identified an overlap between differentially regulated pathways in preeclampsia and cardiovascular system development and function. Cell culture passages 3 and 5 showed similar gene network profiles, and 1260 out of 1266 preeclampsia-associated methylation changes detected in passage 3 were confirmed in passage 5. Conclusion Methylation modification caused by preeclampsia is stable and detectable even in higher cell culture passages. An epigenetically modified endothelial precursor may influence both normal morphogenesis and postnatal vascular repair capacity. Further studies on epigenetic modifications in complicated pregnancies are needed to facilitate development of EPC based therapies for cardiovascular alterations.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hanover, Germany
| | - Tristan Zindler
- Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hanover, Germany
| | | | | | | | - Helge Frieling
- Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hanover, Germany
| | - Carl A Hubel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Thilo Dörk
- Department of Obstetrics and Gynecology, Hannover Medical School, Hanover, Germany
| | | |
Collapse
|
18
|
von Versen-Höynck F, Narasimhan P, Tierney ESS, Martinez N, Conrad KP, Baker VL, Winn VD. Absent or Excessive Corpus Luteum Number Is Associated With Altered Maternal Vascular Health in Early Pregnancy. Hypertension 2019; 73:680-690. [PMID: 30636549 PMCID: PMC6378337 DOI: 10.1161/hypertensionaha.118.12046] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/05/2018] [Indexed: 01/02/2023]
Abstract
Identifying modifiable factors that contribute to preeclampsia risk associated with assisted reproduction can improve maternal health. Vascular dysfunction predates clinical presentation of preeclampsia. Therefore, we examined if a nonphysiological hormonal milieu, a modifiable state, affects maternal vascular health in early pregnancy. Blood pressure, endothelial function, circulating endothelial progenitor cell numbers, lipid levels, and corpus luteum (CL) hormones were compared in a prospective cohort of women with infertility history based on number of CL: 0 CL (programmed frozen embryo transfer [FET], N=18); 1 CL (spontaneous conception [N=16] and natural cycle FET [N=12]); or >3 CL associated with in vitro fertilization [N=11]. Women with 0 or >3 CL lacked the drop in mean arterial blood pressure compared with those with 1 CL (both P=0.05). Reactive hyperemia index was impaired in women with 0 CL compared with 1 CL ( P=0.04) while baseline pulse wave amplitude was higher with > 3 CL compared with 1 CL ( P=0.01) or 0 CL ( P=0.01). Comparing only FET cycles, a lower reactive hyperemia index and a higher augmentation index is noted in FETs with suppressed CL compared with FETs in a natural cycle (both P=0.03). The number of angiogenic and nonangiogenic circulating endothelial progenitor cell numbers was lower in the absence of a CL in FETs ( P=0.01 and P=0.03). Vascular health in early pregnancy is altered in women with aberrant numbers of CL (0 or >3) and might represent insufficient cardiovascular adaptation contributing to an increased risk of preeclampsia.
Collapse
Affiliation(s)
- Frauke von Versen-Höynck
- Stanford University Medical Center, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, 1195 West Fremont Avenue, Sunnyvale, CA 94087, United States of America
- Hannover Medical School, Department of Obstetrics and Gynecology, Lower Saxony, Germany
| | - Purnima Narasimhan
- Stanford University Medical Center, Department of Obstetrics and Gynecology, 300 Pasteur Drive, HG332, Stanford CA 94035
| | - Elif Seda Selamet Tierney
- Lucile Packard Children’s Hospital, Department of Pediatrics, Division of Pediatric Cardiology, Stanford University, Palo Alto, USA, 750 Welch Road, Suite 325, Heart Center
| | - Nadine Martinez
- Stanford University Medical Center, Department of Obstetrics and Gynecology, 300 Pasteur Drive, HG332, Stanford CA 94035
| | - Kirk P Conrad
- Departments of Physiology and Functional Genomics and Obstetrics and Gynecology, D. H. Barron Reproductive and Perinatal Biology Research Program, University of Florida College of Medicine, Gainesville, FL 32610, United States of America
| | - Valerie L. Baker
- Hannover Medical School, Department of Obstetrics and Gynecology, Lower Saxony, Germany
| | - Virginia D Winn
- Stanford University Medical Center, Department of Obstetrics and Gynecology, 300 Pasteur Drive, HG332, Stanford CA 94035
| |
Collapse
|
19
|
Wang Y, Lim R, Nie G. HtrA4 may play a major role in inhibiting endothelial repair in pregnancy complication preeclampsia. Sci Rep 2019; 9:2728. [PMID: 30804477 PMCID: PMC6389976 DOI: 10.1038/s41598-019-39565-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/15/2019] [Indexed: 12/03/2022] Open
Abstract
Preeclampsia (PE) is a life-threatening complication of human pregnancy with no effective treatment other than premature delivery. It is hallmarked by systemic endothelial injury/dysfunction which is believed to be caused by abnormal levels/types of placenta-derived factors that are circulating in the maternal blood. Emerging evidence suggests that endothelial repair is also dysregulated in PE, as circulating endothelial progenitor cells (EPCs) critical for endothelial regeneration are reduced in number and functionality. However, the underlying mechanisms are poorly understood. HtrA4 is a placenta-specific protease that is secreted into the circulation and significantly elevated in early-onset PE. Here we investigated the impact of HtrA4 on endothelial proliferation and repair. We demonstrated that high levels of HtrA4 halted endothelial cell proliferation and significantly down-regulated a number of genes that are critical for cell cycle progression, including CDKN3, BIRC5, CDK1 and MKI67. Furthermore, HtrA4 significantly inhibited the proliferation of primary EPCs isolated from term human umbilical cord blood and impeded their differentiation into mature endothelial cells. Our data thus suggests that elevated levels of HtrA4 in the early-onset PE circulation may impair endothelial cell repair, not only by halting endothelial cell proliferation, but also by inhibiting the proliferation and differentiation of circulating EPCs.
Collapse
Affiliation(s)
- Yao Wang
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3168, Australia
| | - Guiying Nie
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia. .,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia. .,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
20
|
Montazersaheb S, Kabiri F, Saliani N, Nourazarian A, Avci ÇB, Rahbarghazi R, Nozad Charoudeh H. Prolonged incubation with Metformin decreased angiogenic potential in human bone marrow mesenchymal stem cells. Biomed Pharmacother 2018; 108:1328-1337. [PMID: 30372835 DOI: 10.1016/j.biopha.2018.09.135] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/15/2022] Open
Abstract
Metformin is commonly prescribed as a hypoglycemic agent following the onset of type 2 diabetes mellitus. This study aimed to investigate pro- and/or anti-angiogenic effects of Metformin on human bone marrow mesenchymal stem cells. Cells were incubated with different doses of Metformin including 0.5, 1, 10, 50, 100, 200 and 500 μM for 14 days. Cell viability and total fatty acids profile were examined by MTT and gas chromatography methods. Differentiation of cells to endothelial lineage was studied by monitoring the expression of VEGFR-2 and Tie-2 receptors and VE-cadherin via real-time PCR and western blotting. Angiogenic potential and migration of cells were assessed by tubulogenesis and Transwell migration assays. PCR array was performed to analyze mTOR signaling. CD133+ and VEGFR-2+ cells were detected in blood samples of non-diabetic control, diabetic subjects and diabetics received Metformin. Metformin dose-dependently reduced cell survival. Decreased content of palmitate and oleate coincided increased level of stearate, palmitoleate, and linoleate (p < 0.05). Metformin decreased the angiogenic potential of cells by decreasing VEGFR-2 and Tie-2 expression (p < 0.05). The protein level of VE-cadherin decreased in cells received Metformin. Compared to the control, Metformin blunted the expression of VEGF subtypes and directed cells to energy status by induction of PRKAA1, PRKAB2, and PRKAG1 genes (p < 0.05). Non-significant differences were observed regarding the number of CD133 and VEGFR-2 cells in blood samples (p > 0.05). These data support a notion that Metformin could blunt the angiogenic behavior of human mesenchymal stem cells by modulating mTOR signaling pathway.
Collapse
Affiliation(s)
- Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fahimeh Kabiri
- Department of Developmental Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Negar Saliani
- Department of Cellular and Molecular Biology, School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Çıgır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
21
|
de Moreuil C, Fauchais AL, Merviel P, Tremouilhac C, Le Moigne E, Pasquier E, Pan-Petesch B, Lacut K. [Pre-eclampsia prevention in 2018 in general population and in lupic women: At the dawn of a personalized medicine?]. Rev Med Interne 2018; 39:935-941. [PMID: 29933972 DOI: 10.1016/j.revmed.2018.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/27/2018] [Accepted: 06/03/2018] [Indexed: 11/17/2022]
Abstract
Pre-eclampsia prevention represents a major public health issue, as this vasculo-placental disorder generates a great burden of foeto-maternal morbi-mortality. Aspirin has proved its efficacy in primary and secondary pre-eclampsia prevention, especially when it is given at 150mg per day bedtime before 15 weeks of gestation to high-risk women. In the English trial ASPRE, high-risk women were identified by an algorithm taking into account angiogenic biomarkers ascertained at the end of first trimester of pregnancy. This article focuses on physiopathological mechanisms and risk factors of pre-eclampsia and on the interest of early angiogenic biomarkers dosing during pregnancy, for the assessment of pre-eclampsia risk. Unlike Great Britain or Israel, cost-effectiveness of this algorithm in general population has not been assessed in France. Finally, systemic lupus erythematous is at high risk of vasculo-placental disorders. Although few studies of angiogenic biomarkers dosing during lupus pregnancies identified a correlation between high sFlt1 levels at the end of first trimester and subsequent onset of severe vasculo-placental disorders, with a very good negative predictive value of sFtl1. Angiogenic biomarkers ascertainment for screening of vasculo-placental disorders in pregnant women with systemic lupus erythematous could allow targeting at best women needing an aspirin treatment and a closer monitoring.
Collapse
Affiliation(s)
- C de Moreuil
- Département de médecine interne et pneumologie, hôpital La Cavale Blanche, CHU de Brest, boulevard Tanguy-Prigent, 29609 Brest cedex, France; EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France.
| | - A-L Fauchais
- Département de médecine interne, CHU de Limoges, 29200 Limoges, France
| | - P Merviel
- EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France; Service de gynécologie et d'obstétrique, hôpital Morvan, CHU de Brest, 29200 Brest, France
| | - C Tremouilhac
- EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France; Service de gynécologie et d'obstétrique, hôpital Morvan, CHU de Brest, 29200 Brest, France
| | - E Le Moigne
- Département de médecine interne et pneumologie, hôpital La Cavale Blanche, CHU de Brest, boulevard Tanguy-Prigent, 29609 Brest cedex, France; EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France
| | - E Pasquier
- Département de médecine interne et pneumologie, hôpital La Cavale Blanche, CHU de Brest, boulevard Tanguy-Prigent, 29609 Brest cedex, France; EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France
| | - B Pan-Petesch
- EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France; Fédération de cancérologie et d'hématologie, hôpital Morvan, CHU de Brest, 29200 Brest, France
| | - K Lacut
- Département de médecine interne et pneumologie, hôpital La Cavale Blanche, CHU de Brest, boulevard Tanguy-Prigent, 29609 Brest cedex, France; EA 3878, GETBO, université Bretagne-Loire, 29200 Brest cedex, France
| |
Collapse
|
22
|
Szpera-Goździewicz A, Majcherek M, Boruczkowski M, Goździewicz T, Dworacki G, Wicherek L, Bręborowicz GH. Circulating endothelial cells, circulating endothelial progenitor cells, and von Willebrand factor in pregnancies complicated by hypertensive disorders. Am J Reprod Immunol 2017; 77. [PMID: 28224722 DOI: 10.1111/aji.12625] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022] Open
Abstract
PROBLEM We tested the hypothesis that the number of both CECs and CEPCs as well as the vWf blood plasma concentration are altered in pregnant women with hypertensive disorders. METHOD OF STUDY Seventy-five pregnant women were enrolled in our study. We used multicolor flow cytometry for CEC and CEPC analysis and the commercial human VWF ELISA kit to measure vWf blood plasma concentration. RESULTS The highest number of CECs was found in the chronic hypertension group and the lowest number in the healthy pregnant control group. The highest number of CEPCs was found in the control group and the lowest number in the chronic hypertension group. The vWf blood plasma concentration was the highest in the pre-eclampsia group. The CEPC/CEC ratio reached its lowest value in the chronic hypertension group and its highest value in the control group. CONCLUSION The number of both CECs and CEPCs as well as the vWf blood plasma concentration depends on the type of hypertension complicating the pregnancy.
Collapse
Affiliation(s)
- Agata Szpera-Goździewicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Majcherek
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Boruczkowski
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Goździewicz
- Department of Gynecology, Poznan University of Medical Sciences, Poznan, Poland
| | - Grzegorz Dworacki
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Lukasz Wicherek
- Department of Oncology, Radiotherapy and Gynecological Oncology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | |
Collapse
|
23
|
Circulating progenitor and angiogenic cell frequencies are abnormally static over pregnancy in women with preconception diabetes: A pilot study. PLoS One 2017; 12:e0172988. [PMID: 28278173 PMCID: PMC5344347 DOI: 10.1371/journal.pone.0172988] [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: 09/29/2016] [Accepted: 02/13/2017] [Indexed: 11/30/2022] Open
Abstract
Type 1 and 2 diabetes decrease the frequencies and functional capacities of circulating angiogenic cells (CAC). Diabetes also elevates gestational complications. These observations may be interrelated. We undertook pilot studies to address the hypothesis that preconception diabetes deviates known gestational increases in CACs. Cross-sectional study of type 1 diabetic, type 2 diabetic and normoglycemic pregnant women was conducted at 1st, 2nd, and 3rd trimester and compared to a 6mo postpartum surrogate baseline. Circulating progenitor cells (CPC; CD34+CD45dimSSlow) and CACs (CD34+CD45dimSSlow expressing CD133 without or with KDR) were quantified by flow cytometry and by colony assay (CFU-Hill). In pregnant normoglycemic women, CD34+CD45dimSSlow cell frequency was greater in 1st and 3rd trimester than postpartum but frequency of these cells was static over type 1 or 2 diabetic pregnancies. Type 1 and type 2 diabetic women showed CACs variance versus normal controls. Type 1 diabetic women had more total CD34+KDR+ CACs in 1st trimester and a higher ratio of CD133+KDR+ to total CD133+ cells in 1st and 2nd trimesters than control women, demonstrating an unbalance in CD133+KDR+ CACs. Type 2 diabetic women had more CD133+KDR+ CACs in 1st trimester and fewer CD133+KDR- CACs at mid-late pregnancy than normal pregnant women. Thus, pregnancy stage-specific physiological fluctuation in CPCs (CD34+) and CACs (CD133+KDR+ and CD133+KDR-) did not occur in type 1 and type 2 diabetic women. Early outgrowth colonies were stable across normal and diabetic pregnancies. Therefore, preconception diabetes blocks the normal dynamic pattern of CAC frequencies across gestation but does not alter colony growth. The differences between diabetic and typical women were seen at specific gestational stages that may be critical for initiation of the uterine vascular pathologies characterizing diabetic gestations.
Collapse
|
24
|
Gumina DL, Black CP, Balasubramaniam V, Winn VD, Baker CD. Umbilical Cord Blood Circulating Progenitor Cells and Endothelial Colony-Forming Cells Are Decreased in Preeclampsia. Reprod Sci 2016; 24:1088-1096. [PMID: 27879452 DOI: 10.1177/1933719116678692] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Preeclampsia (PE) is a pregnancy-specific disease characterized by the new onset of hypertension and proteinuria. Mothers with PE are known to develop endothelial dysfunction, but its effect on infants has been understudied, as newborns are often asymptomatic. Recent studies indicate that infants born from preeclamptic pregnancies develop endothelial dysfunction including higher blood pressure during childhood and an increased risk of stroke later in life. We hypothesize that PE reduces the number and function of fetal angiogenic progenitor cells and may contribute to this increased risk. We quantified 2 distinct types of angiogenic progenitors, pro-angiogenic circulating progenitor cells (CPCs) and endothelial colony-forming cells (ECFCs), from the umbilical cord blood of preeclamptic pregnancies and normotensive controls. Pro-angiogenic and nonangiogenic CPCs were enumerated via flow cytometry and ECFCs by cell culture. Additionally, we studied the growth, migration, and tube formation of ECFCs from PE and gestational age-matched normotensive control pregnancies. We found that PE resulted in decreased cord blood pro-angiogenic CPCs and ECFCs. Nonangiogenic CPCs were also decreased. Preeclamptic ECFCs demonstrated decreased growth and migration but formed tube-like structures in vitro similar to controls. Our results suggest that the preeclamptic environment alters the number and function of angiogenic progenitor cells and may increase the risk of later vascular disease.
Collapse
Affiliation(s)
- Diane L Gumina
- 1 Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Claudine P Black
- 2 Department of Pediatrics, Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vivek Balasubramaniam
- 3 Pediatric Pulmonology, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Virginia D Winn
- 1 Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
- 4 Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
- Authors contributed equally to the article
| | - Christopher D Baker
- 2 Department of Pediatrics, Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO, USA
- Authors contributed equally to the article
| |
Collapse
|
25
|
Strong inhibitory effect of pre-eclampsia serum on angiogenesis detected in vitro by human cell-based angiogenesis tests. Pregnancy Hypertens 2016; 6:367-373. [DOI: 10.1016/j.preghy.2016.08.239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/26/2016] [Indexed: 11/21/2022]
|
26
|
Liu X, Luo Q, Zheng Y, Liu X, Hu Y, Liu W, Luo M, Tao H, Wu D, Zhao Y, Zou L. Notch1 Impairs Endothelial Progenitor Cell Bioactivity in Preeclampsia. Reprod Sci 2016; 24:47-56. [PMID: 27189202 DOI: 10.1177/1933719116648411] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aberrant vasculature and endothelial dysfunction on both the maternal and the fetal side are thought to play a role in the pathogenesis of preeclampsia, a hypertensive complication during pregnancy. Endothelial progenitor cells (EPCs) have the capacity for endothelial repair. The Dll4/Notch signaling pathway suppresses the functions of EPCs in the pathogenesis of preeclampsia. Notch1 was found to be one of the specific receptors for ligands of the Delta 4 and play critical roles in angiogenesis. However, the roles of Notch1 with regard to EPCs and preeclampsia have yet to be completely characterized. The aim of this study is to determine whether Notch1 also has a negative influence on the regulation of EPC activity. Accordingly, we analyzed the differences between the preeclampsia group and the control group in terms of the number of EPCs and colony-forming units (CFUs) and their Notch1 expressions. The influence of the Notch1 signaling pathway on functions of EPCs was determined by repeating the assays in the presence of Notch1 downregulation. The number of EPCs and CFUs was significantly lower in patients with preeclampsia compared to healthy controls. Additionally, there was a notable increase in Notch1 expression in EPCs of patients with preeclampsia compared to controls. The downregulation of Notch1 promoted the proliferation, differentiation, migration, and adhesion of EPCs and the ability to form human umbilical vein endothelial cell tubes. These findings suggested that decrease and dysfunction of EPCs may be involved in the pathogenesis of preeclampsia. Inhibition of Notch1, which promoted EPC-mediated angiogenesis in vitro, may be an alternative therapeutic approach to promoting vasculogenesis in patients with preeclampsia.
Collapse
Affiliation(s)
- Xiaoxia Liu
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Luo
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanfang Zheng
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Liu
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Hu
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weifang Liu
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglian Luo
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Tao
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Zhao
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- 1 Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Onoyama S, Qiu L, Low HP, Chang CI, Strohsnitter WC, Norwitz ER, Lopresti M, Edmiston K, Lee IM, Trichopoulos D, Lagiou P, Hsieh CC. Prenatal Maternal Physical Activity and Stem Cells in Umbilical Cord Blood. Med Sci Sports Exerc 2016. [PMID: 26197028 DOI: 10.1249/mss.0000000000000731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Early life processes, through influence on fetal stem cells, affect postnatal and adult health outcomes. This study examines the effects of physical activity before and during pregnancy on stem cell counts in umbilical cord blood. METHODS We isolated mononuclear cells from umbilical cord blood samples from 373 singleton full-term pregnancies and quantified hematopoietic (CD34(+), CD34(+)CD38(-), and CD34(+) c-kit(+)), endothelial (CD34(+)CD133(+), CD34(+)CD133(+)VEGFR2(+), CD34(+)VEGFR2(+), and CD133(+)VEGFR2(+)), and putative breast (EpCAM(+), EpCAM(+)CD49f(+), EpCAM(+)CD49f(+)CD117(+), CD49f(+)CD24(+), CD24(+)CD29(+), and CD24(+)CD29(+)CD49f(+)) stem/progenitor cell subpopulations by flow cytometry. Information on physical activities before and during pregnancy was obtained from questionnaires. Weekly energy expenditure was estimated based on metabolic equivalent task values. RESULTS Prepregnancy vigorous exercise was associated positively with levels of endothelial CD34(+)CD133(+), CD34(+)CD133(+)VEGFR2(+), CD34(+)VEGFR2(+), and CD133(+)VEGFR2(+ )progenitor cell populations (P = 0.02, P = 0.01, P = 0.001, and P = 0.003, respectively); positive associations were observed in samples from the first births and those from the second or later births. Prepregnancy moderate and light exercises and light exercise during the first trimester were not significantly associated with any stem/progenitor cell population. Light exercise during the second trimester was positively associated with CD34(+)VEGFR2(+) endothelial progenitor cells (P = 0.03). In addition, levels of EpCAM(+)CD49f(+) and CD49f(+)CD24(+) breast stem cells were significantly lower among pregnant women who engaged in vigorous/moderate exercise during pregnancy (P = 0.05 and P = 0.02, respectively). CONCLUSIONS Vigorous exercise before pregnancy increases the number of endothelial progenitor cells in umbilical cord blood and thus could potentially enhance endothelial function and improve cardiovascular fitness in the offspring. Findings of lower levels of putative breast stem cell subpopulations could have implications on exercise and breast cancer prevention. Prenatal effects of exercise on fetal stem cells warrant further studies.
Collapse
Affiliation(s)
- Sagano Onoyama
- 1Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA; 2Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA; 3Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School and UMass Memorial Health Care, Worcester, MA; 4Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; 5Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; and 6Department of Hygiene, Epidemiology, and Medical Statistics, University of Athens, Athens, GREECE
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Role of axl in preeclamptic EPCs functions. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2016; 36:395-401. [PMID: 27376810 DOI: 10.1007/s11596-016-1598-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/03/2016] [Indexed: 12/14/2022]
Abstract
Axl encodes the tyrosine-protein kinase receptor, participating in the proliferation and migration of many cells. This study examined the role of Axl in functions of endothelial progenitor cells (EPCs). Axl was detected by RT-PCR and Western blotting in both placentas and EPCs from normal pregnancy and preeclampsia patients. The Axl inhibitor, BMS777-607, was used to inhibit the Axl signalling pathway in EPCs. Cell proliferation, differentiation, migration and adhesion were measured by CCK-8 assay, cell differentiation assay, Transwell assay, and cell adhesion assay, respectively. Results showed the expression levels of Axl mRNA and protein were significantly higher in both placentas and EPCs from preeclampsia patients than from normal pregnancy (P<0.05). After treatment with BMS777-607, proliferation, differentiation, migration and adhesion capability of EPCs were all significantly decreased. Our study suggests Axl may play a role in the function of EPCs, thereby involving in the pathogenesis of preeclampsia.
Collapse
|
29
|
Liu X, Luo Q, Zheng Y, Liu X, Hu Y, Liu W, Luo M, Zhao Y, Zou L. NOTCH4 signaling controls EFNB2-induced endothelial progenitor cell dysfunction in preeclampsia. Reproduction 2016; 152:47-55. [PMID: 27069008 DOI: 10.1530/rep-16-0132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 04/08/2016] [Indexed: 12/13/2022]
Abstract
Preeclampsia is a serious complication of pregnancy and is closely related to endothelial dysfunction, which can be repaired by endothelial progenitor cells (EPCs). The DLL4/NOTCH-EFNB2 (ephrinB2) cascade may be involved in the pathogenesis of preeclampsia by inhibiting the biological activity of EPCs. In addition, both NOTCH1 and NOTCH4, which are specific receptors for DLL4/NOTCH, play critical roles in the various steps of angiogenesis. However, it has not been determined which receptor (NOTCH1, NOTCH4, or both) is specific for the DLL4/NOTCH-EFNB2 cascade. Accordingly, we performed a series of investigations to evaluate it. EFNB2 expression was examined when NOTCH4 or NOTCH1 was downregulated, with or without DLL4 treatment. Then, the effects of NOTCH4 on EPC function were detected. Additionally, we analyzed NOTCH4 and EFNB2 expression in the EPCs from preeclampsia and normal pregnancies. Results showed that NOTCH4 downregulation led to decreased expression of EFNB2, which maintained the same level in the presence of DLL4/NOTCH activation. By contrast, NOTCH1 silencing resulted in a moderate increase in EFNB2 expression, which further increased in the presence of DLL4/NOTCH activation. The downregulation of NOTCH4 resulted in an increase of EPC biological activity, which was similar to EFNB2 silencing. NOTCH4 expression, consistent with the EFNB2 level, increased notably in preeclampsia EPCs compared with the controls. These findings suggest that NOTCH4, not NOTCH1, is the specific receptor for the DLL4/NOTCH-EFNB2 cascade. Blockade of this cascade may enhance the angiogenic property of EPCs, and act as a potential target to promote angiogenesis in patients with preeclampsia.
Collapse
Affiliation(s)
- Xiaoxia Liu
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Luo
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yanfang Zheng
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Liu
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Hu
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Weifang Liu
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Minglian Luo
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Zhao
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and GynecologyUnion Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Flo K, Blix ES, Husebekk A, Thommessen A, Uhre AT, Wilsgaard T, Vårtun Å, Acharya G. A longitudinal study of maternal endothelial function, inflammatory response and uterine artery blood flow during the second half of pregnancy. Acta Obstet Gynecol Scand 2015; 95:225-32. [DOI: 10.1111/aogs.12802] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/07/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Kari Flo
- Women's Health and Perinatology Research Group; Department of Clinical Medicine; Faculty of Health Sciences; UiT- The Arctic University of Norway; Tromsø Norway
- Department of Obstetrics and Gynecology; University Hospital of North Norway; Tromsø Norway
| | - Egil S. Blix
- Department of Oncology; University Hospital of North Norway; Tromsø Norway
- Immunology research group; Department of Medical Biology; UiT- The Arctic University of Norway; Tromsø Norway
| | - Anne Husebekk
- Immunology research group; Department of Medical Biology; UiT- The Arctic University of Norway; Tromsø Norway
| | - Anders Thommessen
- Immunology research group; Department of Medical Biology; UiT- The Arctic University of Norway; Tromsø Norway
| | - Andreas T. Uhre
- Immunology research group; Department of Medical Biology; UiT- The Arctic University of Norway; Tromsø Norway
| | - Tom Wilsgaard
- Department of Community Medicine; Faculty of Health Sciences; UiT- The Arctic University of Norway; Tromsø Norway
| | - Åse Vårtun
- Women's Health and Perinatology Research Group; Department of Clinical Medicine; Faculty of Health Sciences; UiT- The Arctic University of Norway; Tromsø Norway
| | - Ganesh Acharya
- Women's Health and Perinatology Research Group; Department of Clinical Medicine; Faculty of Health Sciences; UiT- The Arctic University of Norway; Tromsø Norway
- Department of Obstetrics and Gynecology; University Hospital of North Norway; Tromsø Norway
| |
Collapse
|
31
|
Liu X, Luo Q, Zheng Y, Liu X, Hu Y, Wang F, Zou L. The role of Delta-like 4 ligand/Notch-ephrin-B2 cascade in the pathogenesis of preeclampsia by regulating functions of endothelial progenitor cell. Placenta 2015. [DOI: 10.1016/j.placenta.2015.07.123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
SAHIN HAYRETTIN, GUNEL TUBA, BENIAN ALI, ONAY UCAR EVREN, GURALP ONUR, KILIC AYDINLI. Genomic and proteomic investigation of preeclampsia. Exp Ther Med 2015; 10:711-716. [DOI: 10.3892/etm.2015.2509] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 04/28/2015] [Indexed: 11/06/2022] Open
|
33
|
Brennan LJ, Morton JS, Davidge ST. Vascular dysfunction in preeclampsia. Microcirculation 2014; 21:4-14. [PMID: 23890192 DOI: 10.1111/micc.12079] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/22/2013] [Indexed: 12/30/2022]
Abstract
Preeclampsia is a complex disorder which affects an estimated 5% of all pregnancies worldwide. It is diagnosed by hypertension in the presence of proteinuria after the 20th week of pregnancy and is a prominent cause of maternal morbidity and mortality. As delivery is currently the only known treatment, preeclampsia is also a leading cause of preterm delivery. Preeclampsia is associated with maternal vascular dysfunction, leading to serious cardiovascular risk both during and following pregnancy. Endothelial dysfunction, resulting in increased peripheral resistance, is an integral part of the maternal syndrome. While the cause of preeclampsia remains unknown, placental ischemia resulting from aberrant placentation is a fundamental characteristic of the disorder. Poor placentation is believed to stimulate the release of a number of factors including pro- and antiangiogenic factors and inflammatory activators into the maternal systemic circulation. These factors are critical mediators of vascular function and impact the endothelium in distinctive ways, including enhanced endothelial oxidative stress. The mechanisms of action and the consequences on the maternal vasculature will be discussed in this review.
Collapse
Affiliation(s)
- Lesley J Brennan
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute and the Cardiovascular Research Centre, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
34
|
Beasley KM, Lovering AT, Gilbert JS. Decreased endothelial progenitor cells in preeclampsia and consequences for developmental programming. Hypertension 2014; 64:23-5. [PMID: 24752433 PMCID: PMC4100588 DOI: 10.1161/hypertensionaha.114.03200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kara M Beasley
- From the Department of Human Physiology, University of Oregon, Eugene
| | - Andrew T Lovering
- From the Department of Human Physiology, University of Oregon, Eugene
| | - Jeffrey S Gilbert
- From the Department of Human Physiology, University of Oregon, Eugene.
| |
Collapse
|
35
|
von Versen-Höynck F, Brodowski L, Dechend R, Myerski AC, Hubel CA. Vitamin D antagonizes negative effects of preeclampsia on fetal endothelial colony forming cell number and function. PLoS One 2014; 9:e98990. [PMID: 24892558 PMCID: PMC4044051 DOI: 10.1371/journal.pone.0098990] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/08/2014] [Indexed: 12/22/2022] Open
Abstract
CONTEXT Endothelial dysfunction is a primary feature of preeclampsia, a pregnancy complication associated with an increased future cardiovascular risk for mother and offspring. Endothelial colony forming cells (ECFC) are endothelial progenitor cells that participate in vasculogenesis and endothelial repair. OBJECTIVE We hypothesized that the number and functional properties of fetal cord blood-derived ECFCs are reduced in preeclampsia compared to uncomplicated pregnancy (controls), and asked if adverse effects of preeclampsia on ECFC function are reversed by 1,25 (OH)2 vitamin D3. DESIGN, SETTING, PATIENTS This was a nested, case-control study. Forty women with uncomplicated pregnancy and 33 women with PE were recruited at Magee-Womens Hospital (USA) or at Hannover Medical School (Germany). MAIN OUTCOME MEASURES Time to ECFC colony appearance in culture, and number of colonies formed, were determined. Functional abilities of ECFCs were assessed in vitro by tubule formation in Matrigel assay, migration, and proliferation. ECFC function was tested in the presence or absence of 1,25 (OH)2 vitamin D3, and after vitamin D receptor (VDR) or VEGF signaling blockade. RESULTS The number of cord ECFC colonies was lower (P = 0.04) in preeclampsia compared to controls. ECFCs from preeclampsia showed reduced proliferation (P<0.0001), formed fewer tubules (P = 0.02), and migrated less (P = 0.049) than control. Vitamin D3 significantly improved preeclampsia ECFC functional properties. VDR- or VEGF blockade reduced tubule formation, partially restorable by vitamin D3. CONCLUSION Fetal ECFCs from preeclamptic pregnancies are reduced in number and dysfunctional. Vitamin D3 had rescuing effects. This may have implications for the increased cardiovascular risk associated with preeclampsia.
Collapse
Affiliation(s)
- Frauke von Versen-Höynck
- Department of Obstetrics and Gynecology, Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Lars Brodowski
- Department of Obstetrics and Gynecology, Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (Max-Delbrück Center for Molecular Medicine and Medical Faculty of the Charité and Franz-Volhard Clinic), Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Ashley C. Myerski
- Magee-Womens Research Institute and Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Carl A. Hubel
- Magee-Womens Research Institute and Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
36
|
Brodowski L, Burlakov J, Myerski AC, von Kaisenberg CS, Grundmann M, Hubel CA, von Versen-Höynck F. Vitamin D prevents endothelial progenitor cell dysfunction induced by sera from women with preeclampsia or conditioned media from hypoxic placenta. PLoS One 2014; 9:e98527. [PMID: 24887145 PMCID: PMC4041729 DOI: 10.1371/journal.pone.0098527] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/01/2014] [Indexed: 01/17/2023] Open
Abstract
CONTEXT Placenta-derived circulating factors contribute to the maternal endothelial dysfunction underlying preeclampsia. Endothelial colony forming cells (ECFC), a sub-population of endothelial progenitor cells (EPCs), are thought to be involved in vasculogenesis and endothelial repair. Low vitamin D concentrations are associated with an increased risk for preeclampsia. OBJECTIVE We hypothesized that the function of human fetal ECFCs in culture would be suppressed by exposure to preeclampsia-related factors--preeclampsia serum or hypoxic placental conditioned medium--in a fashion reversed by vitamin D. DESIGN, SETTING, PATIENTS ECFCs were isolated from cord blood of uncomplicated pregnancies and expanded in culture. Uncomplicated pregnancy villous placenta in explant culture were exposed to either 2% (hypoxic), 8% (normoxic) or 21% (hyperoxic) O2 for 48 h, after which the conditioned media (CM) was collected. OUTCOME MEASURES ECFC tubule formation (Matrigel assay) and migration were examined in the presence of either maternal serum from preeclampsia cases or uncomplicated pregnancy controls, or pooled CM, in the presence or absence of 1,25(OH)2 vitamin D3. RESULTS 1,25(OH)2 vitamin D3 reversed the adverse effects of preeclampsia serum or CM from hypoxic placenta on ECFCs capillary-tube formation and migration. Silencing of VDR expression by VDR siRNA, VDR blockade, or VEGF pathway blockade reduced ECFC functional abilities. Effects of VDR or VEGF blockade were partially prevented by vitamin D. CONCLUSION Vitamin D promotes the capillary-like tubule formation and migration of ECFCs in culture, minimizing the negative effects of exposure to preeclampsia-related factors. Further evaluation of the role of vitamin D in ECFC regulation and preeclampsia is warranted.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Jennifer Burlakov
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ashley C. Myerski
- Magee- Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | - Magdalena Grundmann
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Carl A. Hubel
- Magee- Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | |
Collapse
|
37
|
Sipos PI, Rens W, Schlecht H, Fan X, Wareing M, Hayward C, Hubel CA, Bourque S, Baker PN, Davidge ST, Sibley CP, Crocker IP. Uterine vasculature remodeling in human pregnancy involves functional macrochimerism by endothelial colony forming cells of fetal origin. Stem Cells 2014; 31:1363-70. [PMID: 23554274 PMCID: PMC3813980 DOI: 10.1002/stem.1385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/26/2013] [Accepted: 03/04/2013] [Indexed: 12/19/2022]
Abstract
The potency of adult-derived circulating progenitor endothelial colony forming cells (ECFCs) is drastically surpassed by their fetal counterparts. Human pregnancy is associated with robust intensification of blood flow and vascular expansion in the uterus, crucial for placental perfusion and fetal supply. Here, we investigate whether fetal ECFCs transmigrate to maternal bloodstream and home to locations of maternal vasculogenesis, primarily the pregnant uterus. In the first instance, endothelial-like cells, originating from mouse fetuses expressing paternal eGFP, were identified within uterine endothelia. Subsequently, LacZ or enhanced green fluorescent protein (eGFP)-labeled human fetal ECFCs, transplanted into immunodeficient (NOD/SCID) fetuses on D15.5 pregnancy, showed similar integration into the mouse uterus by term. Mature endothelial controls (human umbilical vein endothelial cells), similarly introduced, were unequivocally absent. In humans, SRY was detected in 6 of 12 myometrial microvessels obtained from women delivering male babies. The copy number was calculated at 175 [IQR 149-471] fetal cells per millimeter square endothelium, constituting 12.5% of maternal vessel lumina. Cross-sections of similar human vessels, hybridized for Y-chromosome, positively identified endothelial-associated fetal cells. It appears that through ECFC donation, fetuses assist maternal uterine vascular expansion in pregnancy, potentiating placental perfusion and consequently their own fetal supply. In addition to fetal growth, this cellular mechanism holds implications for materno-fetal immune interactions and long-term maternal vascular health.
Collapse
Affiliation(s)
- Peter I Sipos
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Reduction of maternal circulating endothelial progenitor cells in human pregnancies with intrauterine growth restriction. Placenta 2014; 35:431-6. [PMID: 24819155 DOI: 10.1016/j.placenta.2014.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/10/2014] [Accepted: 04/15/2014] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Circulating endothelial progenitor cells (EPCs) may play a crucial role during pregnancy by sustaining adequate placentation and fetal growth. Unambiguous demonstration of EPC increase during pregnancy has been hampered so far by lack of standardized methods for EPC quantification. In this study we used the currently most accepted phenotype for EPC detection for investigating whether maternal circulating EPCs might increase during normal pregnancy and whether they may fail to increase in pregnancy complicated by idiopathic intrauterine growth restriction (IUGR), a leading cause of perinatal mortality and morbidity characterized by insufficient placental perfusion. METHODS Twenty-one non-pregnant women, 44 women during healthy pregnancy progression (9, 13 and 22 women in the first, second and third trimester, respectively) and 11 with pregnancy complicated by idiopathic IUGR were recruited in a cross-sectional study. EPCs in maternal blood were identified as CD45(dim)/CD34+ / KDR+ cells by flow cytometry. Plasmatic cytokines were measured by ELISA. RESULTS We observed a significant and progressive increase of EPCs in normal pregnancy, yet detectable in early pregnancy but even more pronounced in the third trimester. The increase of EPCs was impaired in IUGR-complicated pregnancies at comparable gestational age. The circulating levels of placental growth-factor and stromal-derived-factor-1 were significantly lower in IUGR than normal pregnancies, possibly contributing to EPC impairment. CONCLUSIONS EPC count in maternal circulation may have a great potential as a novel biomarker for pregnancy monitoring and may represent the target of novel therapeutic strategies designed to prevent adverse pregnancy outcomes often occurring in IUGR.
Collapse
|
39
|
Hromadnikova I, Kotlabova K, Pirkova P, Libalova P, Vernerova Z, Svoboda B, Kucera E. The occurrence of fetal microchimeric cells in endometrial tissues is a very common phenomenon in benign uterine disorders, and the lower prevalence of fetal microchimerism is associated with better uterine cancer prognoses. DNA Cell Biol 2013; 33:40-8. [PMID: 24283364 DOI: 10.1089/dna.2013.2125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This is the first study carried out to describe the role of fetal microchimerism (FM) in the pathogenesis of uterine cancer. The prevalence and concentration of male fetal microchimeric cells (FMCs) were examined in endometrial tissues in relation to subtypes of uterine cancer, and the histological grade and stage of the tumor. FM occurrence was analyzed in relation to risk factors, including hypertension, obesity, type 2 diabetes, dyslipidemia, age at cancer diagnosis, and patient pregnancy history. The prevalence and concentration of FMCs were examined in endometrial tissues using real-time polymerase chain reaction, SRY and β-globin sequences as markers for male fetal FMCs and total DNA. The studied group involved 47 type 1 endometrial cancers, 28 type 2 endometrial cancers, and 41 benign uterine diseases. While the prevalence of FM was decreased only in type 1 endometrial cancer, compared with benign uterine disorders (38.3% vs.70.7%; odds ratio [OR]=0.257, 95% confidence interval [CI]: 0.105 to 0.628, p=0.003), FMC concentrations did not differ within examined groups. The lower FM prevalence was detected in low-grade (grade 1 and grade 2) endometrioid cancer (38.3% vs. 70.7%, OR=0.256, 95% CI: 0.105 to 0.627, p=0.003) and in FIGO 1 tumors (40.7% vs. 70.7%, OR=0.285, 95% CI: 0.120 to 0.675, p=0.004). No correlation between FM prevalence or FMC concentrations and risk factors was demonstrated. A lower prevalence of male FM seemed to be associated with better prognoses in uterine cancer based on tumor subtype, histological grade, and stage of the tumor.
Collapse
Affiliation(s)
- Ilona Hromadnikova
- 1 Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University in Prague , Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
40
|
Yan T, Liu Y, Cui K, Hu B, Wang F, Zou L. MicroRNA-126 regulates EPCs function: Implications for a role of miR-126 in preeclampsia. J Cell Biochem 2013; 114:2148-59. [PMID: 23553946 DOI: 10.1002/jcb.24563] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 03/26/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Ting Yan
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| | - Yan Liu
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| | - Kai Cui
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| | - Bin Hu
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| | - Fang Wang
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| | - Li Zou
- Department of Gynecology and Obstetrics; Union Hospital, Huazhong University of Science and Technology; Wuhan; 430022; China
| |
Collapse
|
41
|
Murphy MS, Casselman RC, Smith GN. Postpartum alterations in circulating endothelial progenitor cells in women with a history of pre-eclampsia. Pregnancy Hypertens 2013; 3:178-85. [DOI: 10.1016/j.preghy.2013.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/06/2013] [Indexed: 11/26/2022]
|
42
|
Crocker IP, Sipos PI. Review: Endothelial progenitor cells in pregnancy and obstetric pathologies. Placenta 2013; 34 Suppl:S62-7. [PMID: 23395298 DOI: 10.1016/j.placenta.2013.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/21/2012] [Accepted: 01/03/2013] [Indexed: 01/10/2023]
Abstract
Since their discovery, endothelial progenitor cells (EPCs) have generated considerable interest in vascular biology. They are a heterogeneous population of cells that exist in both the fetus and adult, and are mobilized to support de novo vessel formation or encourage vascular health. This review summarizes our understanding of these cells in pregnancy, paying particular attention to their physiological role in placental development and the uterus, alongside their involvement in related obstetric pathologies.
Collapse
Affiliation(s)
- I P Crocker
- Faculty of Medical and Human Sciences, University of Manchester, Maternal and Fetal Health Research Centre, Institute of Human Development, Manchester, UK.
| | | |
Collapse
|
43
|
El-Badri NS, Groer M. Gender differences in stem cell population are induced by pregnancy. Med Hypotheses 2012; 79:481-3. [DOI: 10.1016/j.mehy.2012.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/26/2012] [Indexed: 12/14/2022]
|
44
|
Abstract
Preeclampsia is an important obstetric complication that arises in 5% of women after the 20(th) week of gestation, for which there is no specific therapy and no cure. Although much of the recent investigation in this field has focused on soluble forms of the angiogenic membrane receptor tyrosine kinase Flt1 and the transforming growth factor β co-receptor Endoglin, there is significant clinical potential for several GPCR targets and their agonists or antagonists in preeclampsia. In this review, we discuss several of the most promising candidates in this category, including calcitonin receptor-like receptor / receptor activity modifying protein 1 complexes, the angiotensin AT1, 2 and Mas receptors, and the relaxin receptor RXFP1. We also address some of the controversies surrounding the roles and therapeutic potential of these GPCRs and their (ant)agonists in preeclampsia.
Collapse
Affiliation(s)
- Jt McGuane
- D.H. Barron Reproductive and Perinatal Biology Outcomes Research Program, and Department of Physiology and Functional Genomics, and of Obstetrics and Gynecology, University of Florida College of Medicine, Gainesville, FL 32610
| | | |
Collapse
|
45
|
Monga R, Buck S, Sharma P, Thomas R, Chouthai NS. Effect of preeclampsia and intrauterine growth restriction on endothelial progenitor cells in human umbilical cord blood. J Matern Fetal Neonatal Med 2012; 25:2385-9. [PMID: 22640270 DOI: 10.3109/14767058.2012.697228] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Endothelial progenitor cells (EPCs) might play important roles in vascular homeostasis. This study evaluated the influence of prematurity, preeclampsia (PE) and intrauterine growth restriction (IUGR) on the EPC population in human umbilical cord blood (CB). METHODS CB was obtained from 19 preterm and 27 term deliveries. Mononuclear cells were isolated by gradient centrifugation and subjected to flow cytometry to obtain percentages of CD45(d) CD34+, CD45(d) CD133+, CD 45(d) CD34+ CD133+, and CD45(d) CD34+ CD133+ VEGFR-2+ cells. Clinical data were obtained using chart review. RESULTS Percentages of EPCs were comparable between preterm and term cord blood. CD45(d) CD34+ CD133+ cells were significantly decreased in CB samples obtained from women with PE (n = 14) [0.01, (0.00-2.6), [median, (range)], as compared to those without PE (n = 32) [1.74 (0.00-3.1)] (p = 0.005). CD45(d) CD133+ CD34- cells were significantly increased in presence of PE [0.43, (0.06-1.38)], (p = 0.002). CD45(d) CD34+ CD133+ cells were significantly decreased in presence of IUGR, with no change in CD45(d) CD133+ CD34- cells. Differences in EPC types associated with PE and IUGR were present only in term CB. CONCLUSIONS Exposure to PE and IUGR is associated with significant changes in EPC population. Future studies are needed to explore the clinical impact of observed changes.
Collapse
Affiliation(s)
- Ranjan Monga
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Children's Hospital of Michigan, Hutzel Women's Hospital, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
46
|
Abdul Wahid FS, Nasaruddin MZ, Mohd Idris MR, Tusimin M, Tumian NR, Abdullah Mahdy Z. Effects of preeclampsia on the yield of hematopoietic stem cells obtained from umbilical cord blood at delivery. J Obstet Gynaecol Res 2012; 38:490-7. [DOI: 10.1111/j.1447-0756.2011.01740.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Siddiqui N, Hladunewich M. Understanding the Link Between the Placenta and Future Cardiovascular Disease. Trends Cardiovasc Med 2011; 21:188-93. [DOI: 10.1016/j.tcm.2012.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
48
|
Gammill HS, Adams Waldorf KM, Aydelotte TM, Lucas J, Leisenring WM, Lambert NC, Nelson JL. Pregnancy, microchimerism, and the maternal grandmother. PLoS One 2011; 6:e24101. [PMID: 21912617 PMCID: PMC3166068 DOI: 10.1371/journal.pone.0024101] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/31/2011] [Indexed: 01/09/2023] Open
Abstract
Background A woman of reproductive age often harbors a small number of foreign cells, referred to as microchimerism: a preexisting population of cells acquired during fetal life from her own mother, and newly acquired populations from her pregnancies. An intriguing question is whether the population of cells from her own mother can influence either maternal health during pregnancy and/or the next generation (grandchildren). Methodology/Principal Findings Microchimerism from a woman's (i.e. proband's) own mother (mother-of-the-proband, MP) was studied in peripheral blood samples from women followed longitudinally during pregnancy who were confirmed to have uncomplicated obstetric outcomes. Women with preeclampsia were studied at the time of diagnosis and comparison made to women with healthy pregnancies matched for parity and gestational age. Participants and family members were HLA-genotyped for DRB1, DQA1, and DQB1 loci. An HLA polymorphism unique to the woman's mother was identified, and a panel of HLA-specific quantitative PCR assays was employed to identify and quantify microchimerism. Microchimerism from the MP was identified during normal, uncomplicated pregnancy, with a peak concentration in the third trimester. The likelihood of detection increased with advancing gestational age. For each advancing trimester, there was a 12.7-fold increase in the probability of detecting microchimerism relative to the prior trimester, 95% confidence intervals 3.2, 50.3, p<0.001. None of the women with preeclampsia, compared with 30% of matched healthy women, had microchimerism (p = 0.03). Conclusions/Significance These results show that microchimerism from a woman's own mother is detectable in normal pregnancy and diminished in preeclampsia, supporting the previously unexplored hypothesis that MP microchimerism may be a marker reflecting healthy maternal adaptation to pregnancy.
Collapse
Affiliation(s)
- Hilary S Gammill
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America.
| | | | | | | | | | | | | |
Collapse
|
49
|
Penno G, Pucci L, Lucchesi D, Lencioni C, Iorio MC, Vanacore R, Storti E, Resi V, Di Cianni G, Del Prato S. Circulating endothelial progenitor cells in women with gestational alterations of glucose tolerance. Diab Vasc Dis Res 2011; 8:202-10. [PMID: 21653675 DOI: 10.1177/1479164111408938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Endothelial progenitor cells (EPCs) play a role in angiogenesis during pregnancy. The aim of this study was to evaluate circulating EPCs in pregnant women with gestational alterations of glucose tolerance. Glucose tolerance, insulin sensitivity and β-cell function were derived from oral glucose tolerance tests in 23 women with normal glucose tolerance (NGT), 18 with gestational impaired glucose tolerance (GIGT) and 24 with gestational diabetes mellitus (GDM). Circulating cells expressing CD34 in combination with CD133, kinase insert domain receptor (KDR) or both were quantified by flow cytometry. Women with GIGT and GDM had lower CD34(+)KDR(+) and CD34(+)CD133( +)KDR(+) cells at 27±3.2 weeks' gestation compared with NGT (ANOVA p<0.02 for both). CD34(+)KDR(+) and CD34(+)CD133(+)KDR(+) cells were inversely correlated with the area-under-the-glucose-curve (p<0.005, for both) and positively to insulin secretion-sensitivity index (p<0.05, for both). Alterations of glucose tolerance during pregnancy are associated with a decrease in EPCs. Hyperglycaemia might exert a direct effect on depletion of EPCs.
Collapse
|
50
|
Acosta JC, Haas DM, Saha CK, Dimeglio LA, Ingram DA, Haneline LS. Gestational diabetes mellitus alters maternal and neonatal circulating endothelial progenitor cell subsets. Am J Obstet Gynecol 2011; 204:254.e8-254.e15. [PMID: 21167470 PMCID: PMC3057499 DOI: 10.1016/j.ajog.2010.10.913] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 09/27/2010] [Accepted: 10/21/2010] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The purpose of this study was to examine whether women with gestational diabetes mellitus (GDM) and their offspring have reduced endothelial progenitor cell subsets and vascular reactivity. STUDY DESIGN Women with GDM, healthy control subjects, and their infants participated. Maternal blood and cord blood were assessed for colony-forming unit-endothelial cells and endothelial progenitor cell subsets with the use of polychromatic flow cytometry. Cord blood endothelial colony-forming cells were enumerated. Vascular reactivity was tested by laser Doppler imaging. RESULTS Women with GDM had fewer CD34, CD133, CD45, and CD31 cells (circulating progenitor cells [CPCs]) at 24-32 weeks' gestation and 1-2 days after delivery, compared with control subjects. No differences were detected in colony-forming unit-endothelial cells or colony-forming unit-endothelial cells. In control subjects, CPCs were higher in the third trimester, compared with the postpartum period. Cord blood from GDM pregnancies had reduced CPCs. Vascular reactivity was not different between GDM and control subjects. CONCLUSION The normal physiologic increase in CPCs during pregnancy is impaired in women with GDM, which may contribute to endothelial dysfunction and GDM-associated morbidities.
Collapse
Affiliation(s)
- Juan C Acosta
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|