1
|
Ortega MA, Pekarek T, De Leon-Oliva D, Boaru DL, Fraile-Martinez O, García-Montero C, Bujan J, Pekarek L, Barrena-Blázquez S, Gragera R, Rodríguez-Benitez P, Hernández-Fernández M, López-González L, Díaz-Pedrero R, Asúnsolo Á, Álvarez-Mon M, García-Honduvilla N, Saez MA, De León-Luis JA, Bravo C. Placental Tissue Calcification and Its Molecular Pathways in Female Patients with Late-Onset Preeclampsia. Biomolecules 2024; 14:1237. [PMID: 39456171 PMCID: PMC11506500 DOI: 10.3390/biom14101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/20/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Preeclampsia (PE) is a complex multisystem disease characterized by hypertension of sudden onset (>20 weeks' gestation) coupled with the presence of at least one additional complication, such as proteinuria, maternal organ dysfunction, or uteroplacental dysfunction. Hypertensive states during pregnancy carry life-threatening risks for both mother and baby. The pathogenesis of PE develops due to a dysfunctional placenta with aberrant architecture that releases factors contributing to endothelial dysfunction, an antiangiogenic state, increased oxidative stress, and maternal inflammatory responses. Previous studies have shown a correlation between grade 3 placental calcifications and an elevated risk of developing PE at term. However, little is known about the molecular pathways leading to placental calcification. In this work, we studied the gene and protein expression of c-Jun N-terminal kinase (JNK), Runt-related transcription factor 2 (RUNX2), osteocalcin (OSC), osteopontin (OSP), pigment epithelium-derived factor (PEDF), MSX-2/HOX8, SOX-9, WNT-1, and β-catenin in placental tissue from women with late-onset PE (LO-PE). In addition, we employed von Kossa staining to detect mineral deposits in placental tissues. Our results show a significant increase of all these components in placentas from women with LO-PE. Therefore, our study suggests that LO-PE may be associated with the activation of molecular pathways of placental calcification. These results could be the starting point for future research to describe the molecular mechanisms that promote placental calcification in PE and the development of therapeutic strategies directed against it.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Tatiana Pekarek
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Julia Bujan
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Silvestra Barrena-Blázquez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Raquel Gragera
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
| | - Patrocinio Rodríguez-Benitez
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.R.-B.); (J.A.D.L.-L.); (C.B.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
- Department of Nephrology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
| | - Mauricio Hernández-Fernández
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Raul Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (D.D.L.-O.); (D.L.B.); (O.F.-M.); (C.G.-M.); (J.B.); (L.P.); (R.G.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.); (Á.A.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Juan A. De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.R.-B.); (J.A.D.L.-L.); (C.B.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.R.-B.); (J.A.D.L.-L.); (C.B.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| |
Collapse
|
2
|
Zafaranieh S, Siwetz M, Leopold-Posch B, Kummer D, Huppertz B, Desoye G, van Poppel M. Placental structural adaptation to maternal physical activity and sedentary behavior: findings of the DALI lifestyle study. Hum Reprod 2024; 39:deae090. [PMID: 38733100 PMCID: PMC11776022 DOI: 10.1093/humrep/deae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/09/2024] [Indexed: 05/13/2024] Open
Abstract
STUDY QUESTION Are maternal levels of moderate-to-vigorous physical activity (MVPA) and sedentary time (ST) in obese pregnant women associated with placental structural adaptations for facilitating oxygen delivery to the fetus? SUMMARY ANSWER Higher maternal MVPA and ST are associated with a higher density of villi, a proxy measure of placental surface area for oxygen delivery to the fetus, without further added placental vessels. WHAT IS KNOWN ALREADY Physical activity during pregnancy intermittently reduces uterine blood flow, potentially limiting placental and fetal oxygen supply. The placenta can mount several adaptive responses, including enlargement of the surface area of villi and/or feto-placental vessels to accommodate fetal needs. Early research on the morphology and growth of the placenta with exercise interventions has shown inconsistencies and is lacking, particularly in non-lean pregnant women. STUDY DESIGN, SIZE, DURATION This study is a secondary longitudinal analysis of the vitamin D and lifestyle intervention for gestational diabetes prevention (DALI) randomized controlled trial. The prospective study was conducted between 2012 and 2015 in nine European countries at 11 different sites. In this analysis, 92 pregnant women with a BMI ≥ 29 kg/m2 were combined into one cohort. PARTICIPANTS/MATERIALS, SETTING, METHODS MVPA and percentage of time spent sedentary (% ST) were measured with accelerometers during gestation. Placental sections were immunostained for endothelial cell-specific CD34. Artificial intelligence (AI)-based stereology assessed villous density, number, and cross-sectional area of vessels on whole-slide images and in selected regions comprising peripheral villi only, where the majority of vascular adaptations occur. Expression of pro- and anti-angiogenic factors was quantified using molecular counting analysis. MAIN RESULTS AND THE ROLE OF CHANCE In multivariable regression, higher levels of maternal MVPA (min/day) were associated with a higher density of villi in both whole-slide images (beta 0.12; 95% CI 0.05, 0.2) and selected regions (0.17; CI 0.07, 0.26). Unexpectedly, ST was also positively associated with density of villi (0.23; CI 0.04, 0.43). MVPA and ST were not associated with vessel count/mm2 villous area, vessel area, or pro- and anti-angiogenic factor mRNA expression. All estimates and statistical significance of the sensitivity analyses excluding smokers, women who developed gestational diabetes or pre-eclampsia and/or pregnancy-induced hypertension were similar in the main analysis. LIMITATIONS, REASONS FOR CAUTION The placenta is a complex organ undergoing dynamic changes. While various adjustments were made to account for different maternal contributing factors, in addition to the outcome measures, various other factors could impact oxygen delivery to the fetus. WIDER IMPLICATIONS OF THE FINDINGS For the first time, we evaluated the association between placental structures quantified using an AI-based approach with objectively measured physical activity and ST at multiple time points in pregnant women with obesity. The observed adaptations contribute to the advancement of our understanding of the hemodynamics and adaptations of the placental unit in response to MVPA and ST. However, our results might not be generalizable to lean pregnant women. STUDY FUNDING/COMPETING INTEREST(S) The DALI project has received funding from the European Community's 7th Framework Program (FP7/2007-2013) under grant agreement no. 242187. The funders had no role in study design, collection of data, analyses, writing of the article, or the decision to submit it for publication. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER ISRCTN70595832.
Collapse
Affiliation(s)
- Saghi Zafaranieh
- Department of Human Movement Science, Sport and Health, University of
Graz, Graz, Austria
| | - Monika Siwetz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz
Research Center, Medical University of Graz, Graz, Austria
| | | | - Daniel Kummer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz
Research Center, Medical University of Graz, Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz
Research Center, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of
Graz, Graz, Austria
| | - Mireille van Poppel
- Department of Human Movement Science, Sport and Health, University of
Graz, Graz, Austria
| |
Collapse
|
3
|
Qi W, Zhu D, Yin P, Gu X, Zhao Z, Li M, Dong C, Tang Q, Xie W, Zhou T, Xia H, Qiu X, Yang X, Gao G. Pigment Epithelium-Derived Factor, a Novel Adipokine, Contributes to Gestational Diabetes Mellitus. J Clin Endocrinol Metab 2023; 109:e356-e369. [PMID: 37440564 DOI: 10.1210/clinem/dgad413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
CONTEXT Excessive insulin resistance, inadequate insulin compensation, or both could result in gestational diabetes mellitus (GDM). Levels of pigment epithelium-derived factor (PEDF), a novel adipokine that could induce insulin resistance, are high in patients with obesity and diabetes. However, the impact of PEDF in pregnancy remains unknown. OBJECTIVE This study aimed to elucidate the role of PEDF on insulin resistance and compensatory elevation of insulin levels during normal pregnancy and in patients with GDM. METHODS In this population-based and cohort study, logistic regression analysis was performed to determine the association of PEDF/adiponectin/leptin levels with the risk of developing GDM and to predict postpartum prediabetes. PEDF protein, PEDF transgenic mice, PEDF knockout mice, and PEDF-neutralized antibodies were used to observe changes in insulin resistance and insulin levels with pregnancy. RESULTS Plasma PEDF levels were increased in normal pregnancy and higher in GDM women. Higher PEDF levels were associated with the increased risk of developing GDM and emerged as a significant independent determinant of postpartum prediabetes in GDM women. Mechanistically, in vivo and in vitro experiments revealed that PEDF induced insulin resistance by inhibiting the insulin signaling pathway. CONCLUSION In addition to insulin resistance and upregulated insulin levels in normal pregnancy and GDM, aberrant PEDF levels can serve as a "fingerprint" of metabolic abnormalities during pregnancy. Thus, PEDF is a valuable biomarker but could interfere with the time course for early diagnosis and prognosis of GDM.
Collapse
Affiliation(s)
- Weiwei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Dan Zhu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ping Yin
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhen Zhao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Miaoxin Li
- Center for Precision Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chang Dong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qilong Tang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wanting Xie
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Huimin Xia
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xia Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
- Guangdong Provincial Key Laboratory of Diabetology& Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Liu J, Yao L, Wang Y. Resveratrol alleviates preeclampsia-like symptoms in rats through a mechanism involving the miR-363-3p/PEDF/VEGF axis. Microvasc Res 2023; 146:104451. [PMID: 36368448 DOI: 10.1016/j.mvr.2022.104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/09/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
Preeclampsia (PE) is a hypertension-associated disease, and resveratrol (RES) is a polyphenol recognized to present beneficial effects in cardiovascular disease including hypertension. Recently, attentions have come to the therapeutic effect of RES in PE, but the underlying molecular mechanisms remain largely unknown. This study sought to delineate the mechanistic basis regarding bioinformatically identified miR-363-3p/PEDF/VEGF axis for RES treatment in PE. PE-like symptoms were induced in vivo in Sprague-Dawley rats by intraperitoneal injection with Ng-nitro-L-arginine methyl ester (L-NAME), and hypoxia was induced in vitro in trophoblasts by CoCl2. Accordingly, RES was found to enhance viability, migration, angiogenesis, and to repress the apoptosis of hypoxic trophoblasts in vitro. Furthermore, in vivo experiments noted that RES alleviated placental injury and promoted angiogenesis in rats with PE-like symptoms in vivo by increasing VEGF via promoting miR-363-3p-mediated PEDF suppression. Collectively, RES ameliorates PE by upregulating VEGF through miR-363-3p-mediated PEDF downregulation, the mechanism of which may be of promising significance to augment RES efficacy in PE treatment.
Collapse
Affiliation(s)
- Jun Liu
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China.
| | - Lifeng Yao
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China
| |
Collapse
|
5
|
A Comparative Review of Pregnancy and Cancer and Their Association with Endoplasmic Reticulum Aminopeptidase 1 and 2. Int J Mol Sci 2023; 24:ijms24043454. [PMID: 36834865 PMCID: PMC9965492 DOI: 10.3390/ijms24043454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The fundamental basis of pregnancy and cancer is to determine the fate of the survival or the death of humanity. However, the development of fetuses and tumors share many similarities and differences, making them two sides of the same coin. This review presents an overview of the similarities and differences between pregnancy and cancer. In addition, we will also discuss the critical roles that Endoplasmic Reticulum Aminopeptidase (ERAP) 1 and 2 may play in the immune system, cell migration, and angiogenesis, all of which are essential for fetal and tumor development. Even though the comprehensive understanding of ERAP2 lags that of ERAP1 due to the lack of an animal model, recent studies have shown that both enzymes are associated with an increased risk of several diseases, including pregnancy disorder pre-eclampsia (PE), recurrent miscarriages, and cancer. The exact mechanisms in both pregnancy and cancer need to be elucidated. Therefore, a deeper understanding of ERAP's role in diseases can make it a potential therapeutic target for pregnancy complications and cancer and offer greater insight into its impact on the immune system.
Collapse
|
6
|
Krstic J, Deutsch A, Fuchs J, Gauster M, Gorsek Sparovec T, Hiden U, Krappinger JC, Moser G, Pansy K, Szmyra M, Gold D, Feichtinger J, Huppertz B. (Dis)similarities between the Decidual and Tumor Microenvironment. Biomedicines 2022; 10:1065. [PMID: 35625802 PMCID: PMC9138511 DOI: 10.3390/biomedicines10051065] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 02/05/2023] Open
Abstract
Placenta-specific trophoblast and tumor cells exhibit many common characteristics. Trophoblast cells invade maternal tissues while being tolerated by the maternal immune system. Similarly, tumor cells can invade surrounding tissues and escape the immune system. Importantly, both trophoblast and tumor cells are supported by an abetting microenvironment, which influences invasion, angiogenesis, and immune tolerance/evasion, among others. However, in contrast to tumor cells, the metabolic, proliferative, migrative, and invasive states of trophoblast cells are under tight regulatory control. In this review, we provide an overview of similarities and dissimilarities in regulatory processes that drive trophoblast and tumor cell fate, particularly focusing on the role of the abetting microenvironments.
Collapse
Affiliation(s)
- Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Alexander Deutsch
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
- Division of Biophysics, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Tina Gorsek Sparovec
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julian Christopher Krappinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Katrin Pansy
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Marta Szmyra
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Daniela Gold
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| |
Collapse
|
7
|
Zheng J, Li Y, Sang Y, Xu L, Jin X, Tao Y, Li D, Du M. Pigment epithelium-derived factor, a novel decidual natural killer cells-derived factor, protects decidual stromal cells via anti-inflammation and anti-apoptosis in early pregnancy. Hum Reprod 2021; 35:1537-1552. [PMID: 32544239 DOI: 10.1093/humrep/deaa118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
STUDY QUESTION What is the role of pigment epithelium-derived factor (PEDF) from decidual natural killer (dNK) cells during early pregnancy? SUMMARY ANSWER PEDF from dNK cells limits the lipopolysaccharide (LPS)-induced apoptosis and inflammation of decidual stromal cells (DSCs) to maintain DSCs homoeostasis and immune balance at the maternal-foetal interface during early pregnancy. WHAT IS KNOWN ALREADY dNK cells, which secrete PEDF, play critical roles during pregnancy via a series of key regulators. PEDF, a multifunctional endogenous glycoprotein, exhibits a wide range of biological actions upon angiogenesis, inflammation, metabolic homoeostasis, immunomodulation etc., providing potential clinical applications. STUDY DESIGN, SIZE, DURATION Natural killer (NK) cells from decidua and peripheral blood as well as DSCs isolated from normal pregnancy (NP) during the first trimester (6-10 weeks) and the matched patients suffering recurrent miscarriage (RM) were studied. RNA-sequencing analysis of dNK cells was performed to screen for potential key genes involved in RM. The expression of PEDF in dNK cells in NP and RM was examined. A coculture system with LPS-stimulated DSCs and NK cell supernatants derived from NP or RM was established to explore the regulatory mechanisms of PEDF at the maternal-foetal interface. PARTICIPANTS/MATERIALS, SETTING, METHODS Peripheral blood and decidual tissues were obtained from women with NP (n = 61) and RM (n = 21). The expression levels of PEDF in NK cells and its receptor (PEDFR) on DSCs were analysed using flow cytometry, western blot and immunohistochemistry. Purified peripheral natural killer (pNK) cells were cocultured with DSCs or trophoblast cells or a combination of both cell types, and PEDF expression in pNK cells was then examined by flow cytometry. DSCs were treated with LPS, an outer-membrane component of Gram-negative bacteria, thereby mimicking an enhanced inflammatory status within decidua, and were cocultured with dNK cell supernatants from NP or RM. In the coculture system, plasmids expressing short hairpin RNA were used to silence PEDFR on DSCs and block the PEDF/PEDFR interaction. Inflammatory cytokines and apoptosis of DSCs treated as described above were assessed by flow cytometry. Western blotting was performed, and the specific signal pathway inhibitors were used to determine downstream PEDF/PEDFR signalling in early decidua. MAIN RESULTS AND THE ROLE OF CHANCE Markedly higher RNA (P < 0.001) and protein expression of PEDF (P < 0.01) was detected in normal dNK cells when compared with pNK cells. Compared with pNK cells cultured alone, PEDF expression in pNK cells was elevated after coculture with DSCs (P < 0.01) or trophoblast cells (P < 0.001). The increased pro-inflammatory cytokine, tumour necrosis factor-α and apoptosis of DSCs following LPS stimulation were suppressed by recombinant human PEDF (P < 0.001) or the supernatant of dNK cells derived from NP (P < 0.001). However, these effects were somewhat abrogated when the PEDF/PEDFR interaction was blocked with PEDFR short hairpin sRNA (P < 0.01). Furthermore, dNK cell-derived PEDF protected DSCs from LPS-induced inflammation via inhibition of nuclear factor kappa-B activation, while also protecting DSCs from LPS-induced apoptosis via promotion of extracellular signal-regulated kinase expression. Compared with NP, both significantly decreased PEDF RNA (P < 0.001) and protein expression (P < 0.001) in dNK cells, but not in pNK cells (P > 0.05), were detected in women with RM. PEDFR on DSCs was also decreased within RM compared with that within NP (P < 0.001). As a result, dNK cell-mediated anti-inflammation (P < 0.01) and anti-apoptosis (P < 0.05) for protection of LPS-treated DSCs was attenuated in patients suffering from RM. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION We cannot exclude the possibility that the differences in amounts of PEDF and its receptor in tissue from NP versus RM women could be caused by the miscarriage event in women with RM. Our experiments only involved human samples investigated in vitro. Experiments in animal models and human study cohorts are still needed to confirm these findings and further clarify the role of PEDF-PEDFR in NP and/or RM. WIDER IMPLICATIONS OF THE FINDINGS To the best of our knowledge, this is the first study to demonstrate PEDF expression and function at the maternal-foetal interface in the first trimester, providing further evidence that PEDF exhibits functional diversity and has great potential for clinical application(s). The findings of selectively high expression of PEDF in normal dNK cells and the PEDF-mediated role of dNK cells during NP and RM help to further elucidate the immune mechanisms behind RM. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Basic Research Programme of China (2017YFC1001403 and 2015CB943300), Nature Science Foundation from National Nature Science Foundation of China (NSFC; 31970859, 81630036, 81501334, 91542116, 31570920, 81490744 and 31171437), the Innovation-oriented Science and Technology Grant from NHC Key Laboratory of Reproduction Regulation (CX2017-2), the Programme of Shanghai Academic/Technology Research Leader (17XD1400900) and the Key Project of Shanghai Basic Research from Shanghai Municipal Science and Technology Commission (STCSM; 12JC1401600). None of the authors has any conflict of interest to declare.
Collapse
Affiliation(s)
- Ji Zheng
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yanhong Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yifei Sang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Ling Xu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xueling Jin
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yu Tao
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meirong Du
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Ma H, Jiang S, Du L, Liu J, Xu X, Lu X, Ma L, Zhu H, Wei J, Yu Y. Conditioned medium from primary cytotrophoblasts, primary placenta-derived mesenchymal stem cells, or sub-cultured placental tissue promoted HUVEC angiogenesis in vitro. Stem Cell Res Ther 2021; 12:141. [PMID: 33596987 PMCID: PMC7890636 DOI: 10.1186/s13287-021-02192-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Background As a large capillary network, the human placenta plays an important role throughout pregnancy. Placental vascular development is complex and delicate and involves many types of placental cells, such as trophoblasts, and mesenchymal stem cells. There has been no systematic, comparative study on the roles of these two groups of placental cells and the whole placental tissue in the placental angiogenesis. In this study, primary cytotrophoblasts (CTBs) from early pregnancy and primary human placenta-derived mesenchymal stem cells (hPDMSCs) from different stages of pregnancy were selected as the cell research objects, and full-term placental tissue was selected as the tissue research object to detect the effects of their conditioned medium (CM) on human umbilical vein endothelial cell (HUVEC) angiogenesis. Methods We successfully isolated primary hPDMSCs and CTBs, collected CM from these placental cells and sub-cultured placental tissue, and then evaluated the effects of the CM on a series of angiogenic processes in HUVECs in vitro. Furthermore, we measured the levels of angiogenic factors in the CM of placental cells or tissue by an angiogenesis antibody array. Results The results showed that not only placental cells but also sub-cultured placental tissue, to some extent, promoted HUVEC angiogenesis in vitro by promoting proliferation, adhesion, migration, invasion, and tube formation. We also found that primary placental cells in early pregnancy, whether CTBs or hPDMSCs, played more significant roles than those in full-term pregnancy. Placental cell-derived CM collected at 24 h or 48 h had the best effect, and sub-cultured placental tissue-derived CM collected at 7 days had the best effect among all the different time points. The semiquantitative angiogenesis antibody array showed that 18 of the 43 angiogenic factors had obvious spots in placental cell-derived CM or sub-cultured placental tissue-derived CM, and the levels of 5 factors (including CXCL-5, GRO, IL-6, IL-8, and MCP-1) were the highest in sub-cultured placental tissue-derived CM. Conclusions CM obtained from placental cells (primary CTBs or hPDMSCs) or sub-cultured placental tissue contained proangiogenic factors and promoted HUVEC angiogenesis in vitro. Therefore, our research is helpful to better understand placental angiogenesis regulation and provides theoretical support for the clinical application of placental components, especially sub-cultured placental tissue-derived CM, in vascular tissue engineering and clinical treatments.
Collapse
Affiliation(s)
- Haiying Ma
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Shenglu Jiang
- Department of Pathophysiology, Zhangjiakou University, No.P19, Pingmen Street, Qiaoxi District, Zhangjiakou, 075000, Hebei Province, China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Jinfang Liu
- Department of Basic Medical Sciences, Basic Medical College, Shan Xi University of Traditional Chinese Medicine, No. 89, Section 1, Jinci Road, Taiyuan, 030024, Shanxi Province, China
| | - Xiaoyan Xu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Hua Zhu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Jun Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China. .,Shenyang Engineering Technology R&D Center of Cell Therapy CO.LTD, No. 400-8, Zhihui 2nd Street, Hunnan District, Shenyang, 110169, Liaoning Province, China.
| |
Collapse
|
9
|
Sheibani N, Zaitoun IS, Wang S, Darjatmoko SR, Suscha A, Song YS, Sorenson CM, Shifrin V, Albert DM, Melgar-Asensio I, Kandela I, Henkin J. Inhibition of retinal neovascularization by a PEDF-derived nonapeptide in newborn mice subjected to oxygen-induced ischemic retinopathy. Exp Eye Res 2020; 195:108030. [PMID: 32272114 DOI: 10.1016/j.exer.2020.108030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 01/24/2023]
Abstract
Retinopathy of prematurity (ROP) is a growing cause of lifelong blindness and visual defects as improved neonatal care worldwide increases survival in very-low-birthweight preterm newborns. Advancing ROP is managed by laser surgery or a single intravitreal injection of anti-VEGF, typically at 33-36 weeks gestational age. While newer methods of scanning and telemedicine improve monitoring ROP, the above interventions are more difficult to deliver in developing countries. There is also concern as to laser-induced detachment and adverse developmental effects in newborns of anti-VEGF treatment, spurring a search for alternative means of mitigating ROP. Pigment epithelium-derived factor (PEDF), a potent angiogenesis inhibitor appears late in gestation, is undetected in 25-28 week vitreous, but present at full term. Its absence may contribute to ROP upon transition from high-to-ambient oxygen environment or with intermittent hypoxia. We recently described antiangiogenic PEDF-derived small peptides which inhibit choroidal neovascularization, and suggested that their target may be laminin receptor, 67LR. The latter has been implicated in oxygen-induced ischemic retinopathy (OIR). Here we examined the effect of a nonapeptide, PEDF 336, in a newborn mouse OIR model. Neovascularization was significantly decreased in a dose-responsive manner by single intravitreal (IVT) injections of 1.25-7.5 μg/eye (1.0-6.0 nmol/eye). By contrast, anti-mouse VEGFA164 was only effective at 25 ng/eye, with limited dose-response. Combination of anti-VEGFA164 with PEDF 336 gave only the poorer anti-VEGF response while abrogating the robust inhibition seen with peptide-alone, suggesting a need for VEGF in sensitizing the endothelium to the peptide. VEGF stimulated 67LR presentation on endothelial cells, which was decreased in the presence of PEDF 336. Mouse and rabbit eyes showed no histopathology or inflammation after IVT peptide injection. Thus, PEDF 336 is a potential ROP therapeutic, but is not expected to be beneficial in combination with anti-VEGF.
Collapse
Affiliation(s)
- Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ismail S Zaitoun
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Shoujian Wang
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Soesiawati R Darjatmoko
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Andrew Suscha
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Yong-Seok Song
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Daniel M Albert
- Department of Ophthalmology, Casey Eye Institute, Oregon Health Sciences University, Portland, USA
| | | | - Irawati Kandela
- Center for Developmental Therapeutics, Northwestern University, Evanston, IL, USA
| | - Jack Henkin
- Center for Developmental Therapeutics, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
10
|
Zhang X, Zhang K, Zhang Y. Pigment epithelium‑derived factor facilitates NLRP3 inflammasome activation through downregulating cytidine monophosphate kinase 2: A potential treatment strategy for missed abortion. Int J Mol Med 2020; 45:1436-1446. [PMID: 32323732 PMCID: PMC7138263 DOI: 10.3892/ijmm.2020.4517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/20/2019] [Indexed: 12/17/2022] Open
Abstract
A number of conditions may underlie the occurrence of missed abortion (MA), including inflammation. Pigment epithelium‑derived factor (PEDF) is a novel mediator of the inflammation‑related nucleotide‑binding oligomerization domain‑like receptor protein 3 (NLRP3) inflammasome, which is associated with several human diseases. However, the association between MA and NLRP3 inflammasome, and whether PEDF is reduced in MA, remain unknown. In the present study, the decidua and chorion tissues of patients who had suffered a MA were examined, and a lipopolysaccharide (LPS)‑induced human chorionic trophoblast HTR8/SVneo cell model was established to mimic MA in vitro. The results revealed that cytidine monophosphate kinase 2 (CMPK2) expression and NLRP3 inflammasome activation, downstream pro‑IL‑18 and pro‑IL‑1β expression, and IL‑18 and IL‑1β release, were all significantly increased in MA tissues or LPS‑induced HTR8/SVneo cells. PEDF reversed the increase in CMPK2 expression and activation of the NLRP3 inflammasome axis and, thus, downregulated the production of mitochondrial reactive oxygen species and mitochondrial DNA release, resulting in reduced lactate dehydrogenase release, and a resultant decrease in cell viability. Recovery of CMPK2 expression abolished all the effects of PEDF, indicating that CMPK2 may be an effector downstream of PEDF. PEDF reduced CMPK2 protein levels but did not affect the mRNA levels, and treatment with the proteasomal inhibitor MG132 significantly reversed this reduction in CMPK2 protein levels. Furthermore, a ubiquitination assay of immunoprecipitation demonstrated that CMPK2 was polyubiquitinated in the presence of LPS, PEDF and MG132. These results indicated that the NLRP3 inflammasome is implicated in the pathogenesis of MA, and PEDF may reduce MA through ubiquitin‑dependent proteasomal degradation of CMPK2 to inhibit NLRP3 activation, which may serve as a novel strategy for preventing or reducing the risk of MA.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Gynecology and Obstetrics, Changning Maternity and Infant Health Hospital, Shanghai 200050, P.R. China
| | - Kun Zhang
- Department of Pediatrics, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Yi Zhang
- Department of Gynecology and Obstetrics, Changning Maternity and Infant Health Hospital, Shanghai 200050, P.R. China
| |
Collapse
|
11
|
Sun H, Sui Z, Wang D, Ba H, Zhao H, Zhang L, Li C. Identification of interactive molecules between antler stem cells and dermal papilla cells using an in vitro co-culture system. J Mol Histol 2019; 51:15-31. [PMID: 31858326 DOI: 10.1007/s10735-019-09853-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 11/30/2019] [Indexed: 12/25/2022]
|
12
|
Upregulation of VEGF and PEDF in Placentas of Women with Lower Extremity Venous Insufficiency during Pregnancy and Its Implication in Villous Calcification. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5320902. [PMID: 31886225 PMCID: PMC6925773 DOI: 10.1155/2019/5320902] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022]
Abstract
Pregnancy is a period in a woman's life in which changes can occur that affect different physiological processes. Common conditions during this period include vascular changes, such as lower extremity venous insufficiency (VI). This is an observational, analytical, and prospective cohort study in which 114 pregnant women were analyzed, of which 62 were clinically diagnosed with VI. In parallel, 52 control patients without VI (HC) were studied. The aim of this study was to observe changes in angiogenesis and inflammation markers as well as the presence of calcium deposits. The expression of vascular endothelial growth factor (VEGF), transforming growth factor-β (TGF-β), and pigment epithelium-derived factor (PEDF) was analyzed by immunohistochemistry and RT-qPCR. The presence of calcium deposits was revealed using the von Kossa method. In the placentas of mothers with VI, gene expression of VEGF (34.575 [32.380–36.720] VI vs 32.965 [30.580–36.320] HC) and PEDF (25.417 [24.459–27.675] VI vs 24.400 [23.102–30.223] HC) significantly increased, as was protein expression in the placental villi. An increase in calcium deposits was observed in the placentas of women with VI (72.58% VI/53.84% HC). This study revealed the existence of cellular damage in the placental villi of mothers with VI with tissue implications such as increased calcification.
Collapse
|
13
|
Brook N, Brook E, Dharmarajan A, Chan A, Dass CR. The role of pigment epithelium-derived factor in protecting against cellular stress. Free Radic Res 2019; 53:1166-1180. [PMID: 31760841 DOI: 10.1080/10715762.2019.1697809] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Since its discovery as a neurotrophic factor in retinal pigmented epithelium cells in the late 1980s, there has been an increase in understanding of the role that pigment epithelium-derived factor (PEDF) plays in cellular functions. PEDF plays an important role in mediating cellular protection during exposure to oxidative stress and inflammation by preventing stress-induced angiogenesis and apoptosis. PEDF acts to reduce oxidative stress by promoting mitochondrial stability and by regulating the expression of enzymes involved in ROS accumulation and clearance. PEDF protects against the negative effects of oxidative stress by regulating cell survival pathways and the expression of inflammatory and proangiogenic mediators. PEDF-mediated cellular protection may be of clinical importance in diseases characterised by oxidative stress, chronic inflammation and pathological neovascularization, indicating that targeting PEDF may be a potential focus for therapeutic interventions in chronic diseases. In this review, we provide a historical perspective on the discoveries of PEDF interactions and functions, and discuss recent in vitro, in vivo and clinical findings to provide a current summary of the important protective effects following cellular exposure to stress stimuli and future clinical potential of PEDF.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Arun Dharmarajan
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia.,Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley, Australia.,Hollywood Private Hospital, Breast Clinical Trials Unit, Breast Cancer Research Centre-Western Australia, Nedlands, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|
14
|
Sheibani N, Wang S, Darjatmoko SR, Fisk DL, Shahi PK, Pattnaik BR, Sorenson CM, Bhowmick R, Volpert OV, Albert DM, Melgar-Asensio I, Henkin J. Novel anti-angiogenic PEDF-derived small peptides mitigate choroidal neovascularization. Exp Eye Res 2019; 188:107798. [PMID: 31520600 PMCID: PMC7032632 DOI: 10.1016/j.exer.2019.107798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/07/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
Abnormal migration and proliferation of endothelial cells (EC) drive neovascular retinopathies. While anti-VEGF treatment slows progression, pathology is often supported by decrease in intraocular pigment epithelium-derived factor (PEDF), an endogenous inhibitor of angiogenesis. A surface helical 34-mer peptide of PEDF, comprising this activity, is efficacious in animal models of neovascular retina disease but remains impractically large for therapeutic use. We sought smaller fragments within this sequence that mitigate choroidal neovascularization (CNV). Expecting rapid intravitreal (IVT) clearance, we also developed a method to reversibly attach peptides to nano-carriers for extended delivery. Synthetic fragments of 34-mer yielded smaller anti-angiogenic peptides, and N-terminal capping with dicarboxylic acids did not diminish activity. Charge restoration via substitution of an internal aspartate by asparagine improved potency, achieving low nM apoptotic response in VEGF-activated EC. Two optimized peptides (PEDF 335, 8-mer and PEDF 336, 9-mer) were tested in a mouse model of laser-induced CNV. IVT injection of either peptide, 2-5 days before laser treatment, gave significant CNV decrease at day +14 post laser treatment. The 8-mer also decreased CNV, when administered as eye drops. Also examined was a nanoparticle-conjugate (NPC) prodrug of the 9-mer, having positive zeta potential, expected to display longer intraocular residence. This NPC showed extended efficacy, even when injected 14 days before laser treatment. Neither inflammatory cells nor other histopathologic abnormalities were seen in rabbit eyes harvested 14 days following IVT injection of PEDF 336 (>200 μg). No rabbit or mouse eye irritation was observed over 12-17 days of PEDF 335 eye drops (10 mM). Viability was unaffected in 3 retinal and 2 choroidal cell types by PEDF 335 up to 100 μM, PEDF 336 (100 μM) gave slight growth inhibition only in choroidal EC. A small anti-angiogenic PEDF epitope (G-Y-D-L-Y-R-V) was identified, variants (adipic-Sar-Y-N-L-Y-R-V) mitigate CNV, with clinical potential in treating neovascular retinopathy. Their shared active motif, Y - - - R, is found in laminin (Ln) peptide YIGSR, which binds Ln receptor 67LR, a known high-affinity ligand of PEDF 34-mer.
Collapse
Affiliation(s)
- Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Soesiawati R Darjatmoko
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Debra L Fisk
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Pawan K Shahi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bikash R Pattnaik
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Reshma Bhowmick
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Olga V Volpert
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniel M Albert
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Jack Henkin
- Center for Developmental Therapeutics, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
15
|
Outgrowth, proliferation, viability, angiogenesis and phenotype of primary human endothelial cells in different purchasable endothelial culture media: feed wisely. Histochem Cell Biol 2019; 152:377-390. [PMID: 31541300 PMCID: PMC6842357 DOI: 10.1007/s00418-019-01815-2] [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] [Accepted: 09/06/2019] [Indexed: 12/15/2022]
Abstract
Function and dysfunction of endothelial cells are regulated by a multitude of factors. Endothelial cell research often requires in vitro cell culture experiments. Hence, various culture media specifically designed to promote endothelial cell growth are available. These strikingly differ in their composition: complex media contain endothelial cell growth supplement (ECGS), an extract produced of bovine brain with undefined amounts of biologically active compounds, whilst defined media contain selected growth factors in defined concentrations. We here compared the effect of seven purchasable endothelial cell culture media on colony outgrowth, proliferation, viability, in vitro angiogenesis and phenotype of mature primary human endothelial cells using feto-placental endothelial cells isolated from chorionic arteries (fpEC). The effect of media on colony outgrowth was additionally tested on umbilical cord blood-derived endothelial progenitor cells (ECFCs). Outgrowth, purity, proliferation and viability differed between media. Outgrowth of fpEC and ECFCs was best in a defined medium containing EGF, FGF2 and VEGF. By contrast, established fpEC isolations proliferated best in complex media containing ECGS, heparin and ascorbic acid. Also viability of cells was higher in complex media. In vitro angiogenesis was most intense in a defined medium containing the highest number of individual growth factors. FACS analysis of surface markers for endothelial cell subtypes revealed that endothelial phenotype of fpEC was unaffected by media composition. Our data demonstrate the fundamental effect of endothelial cell culture media on primary cell isolation success and behaviour. Whether the composition of supplements is suitable also for individual experiments needs to be tested specifically.
Collapse
|
16
|
McIntyre HD, Catalano P, Zhang C, Desoye G, Mathiesen ER, Damm P. Gestational diabetes mellitus. Nat Rev Dis Primers 2019; 5:47. [PMID: 31296866 DOI: 10.1038/s41572-019-0098-8] [Citation(s) in RCA: 903] [Impact Index Per Article: 150.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Hyperglycaemia that develops during pregnancy and resolves after birth has been recognized for over 50 years, but uniform worldwide consensus is lacking about threshold hyperglycaemic levels that merit a diagnosis of 'gestational diabetes mellitus' (GDM) and thus treatment during pregnancy. GDM is currently the most common medical complication of pregnancy, and prevalence of undiagnosed hyperglycaemia and even overt diabetes in young women is increasing. Maternal overweight and obesity, later age at childbearing, previous history of GDM, family history of type 2 diabetes mellitus and ethnicity are major GDM risk factors. Diagnosis is usually performed using an oral glucose tolerance test (OGTT), although a non-fasting, glucose challenge test (GCT) is used in some parts of the world to screen women for those requiring a full OGTT. Dietary modification and increased physical activity are the primary treatments for GDM, but pharmacotherapy, usually insulin, is used when normoglycaemia is not achieved. Oral hypoglycaemic agents, principally metformin and glibenclamide (glyburide), are also used in some countries. Treatment improves immediate pregnancy outcomes, reducing excess fetal growth and adiposity and pregnancy-related hypertensive disorders. GDM increases the risk of long-term complications, including obesity, impaired glucose metabolism and cardiovascular disease, in both the mother and infant. Optimal management of mother and infant during long-term follow-up remains challenging, with very limited implementation of preventive strategies in most parts of the world.
Collapse
Affiliation(s)
- H David McIntyre
- Mater Research and University of Queensland, Brisbane, Queensland, Australia.
| | - Patrick Catalano
- Mother Infant Research Institute, Department of Obstetrics and Gynecology, Tufts University School of Medicine, Friedman School of Nutrition Science and Policy, Boston, MA, USA
| | - Cuilin Zhang
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Elisabeth R Mathiesen
- Department of Endocrinology, Center for Pregnant Women with Diabetes, Rigshospitalet and The Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Damm
- Department of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet and The Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Kuo CY, Shevchuk M, Opfermann J, Guo T, Santoro M, Fisher JP, Kim PCW. Trophoblast-endothelium signaling involves angiogenesis and apoptosis in a dynamic bioprinted placenta model. Biotechnol Bioeng 2019; 116:181-192. [PMID: 30298908 PMCID: PMC6289739 DOI: 10.1002/bit.26850] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 01/01/2023]
Abstract
Trophoblast invasion and remodeling of the maternal spiral arteries are required for pregnancy success. Aberrant endothelium-trophoblast crosstalk may lead to preeclampsia, a pregnancy complication that has serious effects on both the mother and the baby. However, our understanding of the mechanisms involved in this pathology remains elementary because the current in vitro models cannot describe trophoblast-endothelium interactions under dynamic culture. In this study, we developed a dynamic three-dimensional (3D) placenta model by bioprinting trophoblasts and an endothelialized lumen in a perfusion bioreactor. We found the 3D printed perfusion bioreactor system significantly augmented responses of endothelial cells by encouraging network formations and expressions of angiogenic markers, cluster of differentiation 31 (CD31), matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-9 (MMP9), and vascular endothelial growth factor A (VEGFA). Bioprinting favored colocalization of trophoblasts with endothelial cells, similar to in vivo observations. Additional analysis revealed that trophoblasts reduced the angiogenic responses by reducing network formation and motility rates while inducing apoptosis of endothelial cells. Moreover, the presence of endothelial cells appeared to inhibit trophoblast invasion rates. These results clearly demonstrated the utility and potential of bioprinting and perfusion bioreactor system to model trophoblast-endothelium interactions in vitro. Our bioprinted placenta model represents a crucial step to develop advanced research approach that will expand our understanding and treatment options of preeclampsia and other pregnancy-related pathologies.
Collapse
Affiliation(s)
- Che-Ying Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Mariya Shevchuk
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Justin Opfermann
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
| | - Ting Guo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Marco Santoro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Peter CW Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- School of Medicine and Health Sciences, The George Washington University, Washington, DC
| |
Collapse
|
18
|
Zhang YG, Yang HL, Zhang YP, Ma QL, Long Y, Zheng ZX. Pigment epithelium–derived factor/vascular endothelial growth factor ratio for early prediction of preeclampsia: A prospective multicenter study in China. Pregnancy Hypertens 2018; 14:43-48. [DOI: 10.1016/j.preghy.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/07/2018] [Accepted: 07/26/2018] [Indexed: 02/02/2023]
|
19
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 452] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Stuart TJ, O’Neill K, Condon D, Sasson I, Sen P, Xia Y, Simmons RA. Diet-induced obesity alters the maternal metabolome and early placenta transcriptome and decreases placenta vascularity in the mouse. Biol Reprod 2018; 98:795-809. [PMID: 29360948 PMCID: PMC6454478 DOI: 10.1093/biolre/ioy010] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/19/2017] [Accepted: 01/17/2018] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity is associated with an increased risk of obesity and metabolic disease in offspring. Increasing evidence suggests that the placenta plays an active role in fetal programming. In this study, we used a mouse model of diet-induced obesity to demonstrate that the abnormal metabolic milieu of maternal obesity sets the stage very early in pregnancy by altering the transcriptome of placenta progenitor cells in the preimplantation (trophectoderm [TE]) and early postimplantation (ectoplacental cone [EPC]) placenta precursors, which is associated with later changes in placenta development and function. Sphingolipid metabolism was markedly altered in the plasma of obese dams very early in pregnancy as was expression of genes related to sphingolipid processing in the early placenta. Upregulation of these pathways inhibits angiogenesis and causes endothelial dysfunction. The expression of many other genes related to angiogenesis and vascular development were disrupted in the TE and EPC. Other key changes in the maternal metabolome in obese dams that are likely to influence placenta and fetal development include a marked decrease in myo and chiro-inositol. These early metabolic and gene expression changes may contribute to phenotypic changes in the placenta, as we found that exposure to a high-fat diet decreased placenta microvessel density at both mid and late gestation. This is the first study to demonstrate that maternal obesity alters the transcriptome at the earliest stages of murine placenta development.
Collapse
Affiliation(s)
- Tami J Stuart
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen O’Neill
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Condon
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Issac Sasson
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Payel Sen
- Epigenetics Center, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yunwei Xia
- College of Arts and Sciences, Cornell University, Ithaca, New York, USA
| | - Rebecca A Simmons
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
From Inflammation to Current and Alternative Therapies Involved in Wound Healing. Int J Inflam 2017; 2017:3406215. [PMID: 28811953 PMCID: PMC5547704 DOI: 10.1155/2017/3406215] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/01/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023] Open
Abstract
Wound healing is a complex event that develops in three overlapping phases: inflammatory, proliferative, and remodeling. These phases are distinct in function and histological characteristics. However, they depend on the interaction of cytokines, growth factors, chemokines, and chemical mediators from cells to perform regulatory events. In this article, we will review the pathway in the skin healing cascade, relating the major chemical inflammatory mediators, cellular and molecular, as well as demonstrating the local and systemic factors that interfere in healing and disorders associated with tissue repair deficiency. Finally, we will discuss the current therapeutic interventions in the wounds treatment, and the alternative therapies used as promising results in the development of new products with healing potential.
Collapse
|
22
|
Diabetic nephropathy: serum miR-9 confers a poor prognosis in and is associated with level changes of vascular endothelial growth factor and pigment epithelium-derived factor. Biotechnol Lett 2017; 39:1583-1590. [PMID: 28667418 DOI: 10.1007/s10529-017-2390-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/20/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the relationship between the serum level of miR-9 and the progression of diabetic nephropathy (DN) and related molecular mechanisms. RESULTS Thirty-five healthy subjects and 140 DN patients were divided into five groups: control, DN I-II, DN III, DN IV and DN V. Serum level of miR-9 was measured by real-time qPCR. Serum levels of vascular endothelial growth factor (VEGF), pigment epithelium-derived factor (PEDF) lipids, fasting glucose, insulin, hemoglobin A1c (HBA1c), creatinine, fibrinogen and insulin resistance (HOMA-IR) were also measured. The results show that the levels of miR-9, PEDF and VEGF are increased with DN progression (P < 0.05). miR-9, VEGF and PEDF are independent risk factors of DN (R2 = 0.430). Spearman rank correlation analysis showed that miR-9 level is positively related to the levels of VEGF, PEDF, cholesterol, triglyceride, fasting glucose, fasting insulin, HBA1c, creatinine, fibrinogen and HOMA-IR (P < 0.05). CONCLUSIONS Serum miR-9 is a potential marker for conferring a poor prognosis in DN and associated with the levels of VEGF, PEDF and biochemical indices.
Collapse
|
23
|
GDM alters paracrine regulation of feto-placental angiogenesis via the trophoblast. J Transl Med 2017; 97:409-418. [PMID: 28112754 DOI: 10.1038/labinvest.2016.149] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 01/13/2023] Open
Abstract
Feto-placental angiogenesis and vascular development are tightly regulated by pro- and anti-angiogenic factors. Villous trophoblast may be a major source of these factors. It forms the classical placental barrier between mother and fetus, and is thus exposed to maternal influences as well. Metabolic and hormonal derangements in gestational diabetes mellitus (GDM) affect feto-placental angiogenesis and vascular growth. Here we hypothesized that GDM alters the trophoblast secretome, which will modulate the paracrine regulation of feto-placental angiogenesis. Primary term trophoblasts were isolated from normal (n=6) and GDM (n=6) pregnancies. Trophoblast conditioned medium (CM) was used to investigate paracrine effects of normal and GDM-exposed trophoblasts on feto-placental endothelial cells (fpECs; n=7), using functional assays for 2D network formation, wound healing, chemotaxis, and proliferation. Gene expression of 23 pro- and anti-angiogenic factors was analyzed. Four trophoblast-derived paracrine regulators of angiogenesis were specifically measured in CM. CM from GDM trophoblasts increased 2D network formation of fpEC by 2.4-fold (P<0.001), whereas wound healing was attenuated by 1.8-fold (P=0.02) and chemo-attraction to the CM was reduced by 33±9% (P=0.02). The effect of CM on proliferation was unchanged between normal and GDM trophoblasts. Expression analysis of pro- and anti-angiogenic molecules in normal and GDM trophoblasts revealed significant differences in ANGPT2, HGF, KISS1 and PLGF expression. Analysis of secreted proteins demonstrated reduced pigment epithelium derived factor and tumor necrosis factor-α secretion by GDM trophoblasts. GDM alters the balance of trophoblast derived, angiogenesis modulating paracrine factors. This may contribute to GDM-associated changes in placental angiogenesis and vascular structure.
Collapse
|
24
|
Stangret A, Skoda M, Wnuk A, Pyzlak M, Szukiewicz D. Mild anemia during pregnancy upregulates placental vascularity development. Med Hypotheses 2017; 102:37-40. [PMID: 28478827 DOI: 10.1016/j.mehy.2017.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/05/2017] [Indexed: 11/16/2022]
Abstract
The connection between maternal hematological status and pregnancy outcome has been shown by many independent researchers. Attention was initially focused on the adverse effects of moderate and severe anemia. Interestingly, some studies revealed that mild anemia was associated with optimal fetal development and was not affecting pregnancy outcome. The explanation for this phenomenon became a target for scientists. Hemodilution, physiologic anemia and relative decrease in hemoglobin concentration are the changes observed during pregnancy but they do not explain the reasons for the positive influence of mild anemia on a fetomaternal unit. It is hypothesized that hemodilution facilitates placental perfusion because blood viscosity is reduced. Subsequently, it may lead to a decline in hemoglobin concentration. Anemia from its definition implies decreased oxygen carrying capacity of the blood and can result in hypoxemia and even hypoxia, which is a common factor inducing new blood vessels formation. Therefore, we raised the hypothesis that the lowered hemoglobin concentration during pregnancy may upregulate vascular growth factor receptors expression such as VEGFR-1 (Flt-1) and VEGFR-2 (FLK-1/KDR). Consecutively, increased fetoplacental vasculogenesis and angiogenesis provide further expansion of vascular network development, better placental perfusion and hence neither fetus nor the mother are affected.
Collapse
Affiliation(s)
- A Stangret
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland.
| | - M Skoda
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - A Wnuk
- Chair and Department of Obstetrics, Gynecology, and Oncology, 2nd Faculty of Medical University of Warsaw, Poland
| | - M Pyzlak
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - D Szukiewicz
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| |
Collapse
|
25
|
Metzler VM, de Brot S, Robinson RS, Jeyapalan JN, Rakha E, Walton T, Gardner DS, Lund EF, Whitchurch J, Haigh D, Lochray JM, Robinson BD, Allegrucci C, Fray RG, Persson JL, Ødum N, Miftakhova RR, Rizvanov AA, Hughes IA, Tadokoro-Cuccaro R, Heery DM, Rutland CS, Mongan NP. Androgen dependent mechanisms of pro-angiogenic networks in placental and tumor development. Placenta 2017; 56:79-85. [PMID: 28238455 DOI: 10.1016/j.placenta.2017.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 11/25/2022]
Abstract
The placenta and tumors share important characteristics, including a requirement to establish effective angiogenesis. In the case of the placenta, optimal angiogenesis is required to sustain the blood flow required to maintain a successful pregnancy, whereas in tumors establishing new blood supplies is considered a key step in supporting metastases. Therefore the development of novel angiogenesis inhibitors has been an area of active research in oncology. A subset of the molecular processes regulating angiogenesis are well understood in the context of both early placentation and tumorigenesis. In this review we focus on the well-established role of androgen regulation of angiogenesis in cancer and relate these mechanisms to placental angiogenesis. The physiological actions of androgens are mediated by the androgen receptor (AR), a ligand dependent transcription factor. Androgens and the AR are essential for normal male embryonic development, puberty and lifelong health. Defects in androgen signalling are associated with a diverse range of clinical disorders in men and women including disorders of sex development (DSD), polycystic ovary syndrome in women and many cancers. We summarize the diverse molecular mechanisms of androgen regulation of angiogenesis and infer the potential significance of these pathways to normal and pathogenic placental function. Finally, we offer potential research applications of androgen-targeting molecules developed to treat cancer as investigative tools to help further delineate the role of androgen signalling in placental function and maternal and offspring health in animal models.
Collapse
Affiliation(s)
- Veronika M Metzler
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Simone de Brot
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Robert S Robinson
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jennie N Jeyapalan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emad Rakha
- School of Medicine and Sciences, University of Nottingham, Nottingham City Hospital, NG5 1PB, UK
| | - Thomas Walton
- Department of Urology, Nottingham University Hospitals NHS Trust, NG5 1PB, UK
| | - David S Gardner
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emma F Lund
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | | | - Daisy Haigh
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jack M Lochray
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Brian D Robinson
- Department of Pathology, Weill Cornell Medicine, New York 10065, USA
| | - Cinzia Allegrucci
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Rupert G Fray
- School of Biosciences, University of Nottingham, LE12 5RD, UK
| | - Jenny L Persson
- Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Molecular Biology, Umeå University, Sweden
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Regina R Miftakhova
- Department of Molecular Biology, Umeå University, Sweden; Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Albert A Rizvanov
- Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Hills Rd, Cambridge CB2 0QQ, UK
| | | | - David M Heery
- School of Pharmacy, University of Nottingham, NG7 2TQ, UK
| | - Catrin S Rutland
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK.
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK; Department of Pharmacology, Weill Cornell Medicine, New York 10065, USA.
| |
Collapse
|