1
|
Kostyunina DS, Pakhomov NV, Jouida A, Dillon E, Baugh JA, McLoughlin P. Transcriptomics and proteomics revealed sex differences in human pulmonary microvascular endothelial cells. Physiol Genomics 2024; 56:194-220. [PMID: 38047313 DOI: 10.1152/physiolgenomics.00051.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
Marked sexual dimorphism is displayed in the onset and progression of pulmonary hypertension (PH). Females more commonly develop pulmonary arterial hypertension, yet females with pulmonary arterial hypertension and other types of PH have better survival than males. Pulmonary microvascular endothelial cells play a crucial role in pulmonary vascular remodeling and increased pulmonary vascular resistance in PH. Given this background, we hypothesized that there are sex differences in the pulmonary microvascular endothelium basally and in response to hypoxia that are independent of the sex hormone environment. Human pulmonary microvascular endothelial cells (HPMECs) from healthy male and female donors, cultured under physiological shear stress, were analyzed using RNA sequencing and label-free quantitative proteomics. Gene set enrichment analysis identified a number of sex-different pathways in both normoxia and hypoxia, including pathways that regulate cell proliferation. In vitro, the rate of proliferation in female HPMECs was lower than in male HPMECs, a finding that supports the omics results. Interestingly, thrombospondin-1, an inhibitor of proliferation, was more highly expressed in female cells than in male cells. These results demonstrate, for the first time, important differences between female and male HPMECs that persist in the absence of sex hormone differences and identify novel pathways for further investigation that may contribute to sexual dimorphism in pulmonary hypertensive diseases.NEW & NOTEWORTHY There is marked sexual dimorphism in the development and progression of pulmonary hypertension. We show differences in RNA and protein expression between female and male human pulmonary microvascular endothelial cells grown under conditions of physiological shear stress, which identify sex-different cellular pathways both in normoxia and hypoxia. Importantly, these differences were detected in the absence of sex hormone differences. The pathways identified may provide novel targets for the development of sex-specific therapies.
Collapse
Affiliation(s)
- Daria S Kostyunina
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Nikolai V Pakhomov
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Amina Jouida
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Eugene Dillon
- Conway Institute, University College Dublin, Dublin, Ireland
| | - John A Baugh
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Dubourg V, Schwerdt G, Schreier B, Kopf M, Mildenberger S, Benndorf RA, Gekle M. EGFR activation differentially affects the inflammatory profiles of female human aortic and coronary artery endothelial cells. Sci Rep 2023; 13:22827. [PMID: 38129563 PMCID: PMC10739936 DOI: 10.1038/s41598-023-50148-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Endothelial cells (EC) are key players in vascular function, homeostasis and inflammation. EC show substantial heterogeneity due to inter-individual variability (e.g. sex-differences) and intra-individual differences as they originate from different organs or vessels. This variability may lead to different responsiveness to external stimuli. Here we compared the responsiveness of female human primary EC from the aorta (HAoEC) and coronary arteries (HCAEC) to Epidermal Growth Factor Receptor (EGFR) activation. EGFR is an important signal integration hub for vascular active substances with physiological and pathophysiological relevance. Our transcriptomic analysis suggested that EGFR activation differentially affects the inflammatory profiles of HAoEC and HCAEC, particularly by inducing a HCAEC-driven leukocyte attraction but a downregulation of adhesion molecule and chemoattractant expression in HAoEC. Experimental assessments of selected inflammation markers were performed to validate these predictions and the results confirmed a dual role of EGFR in these cells: its activation initiated an anti-inflammatory response in HAoEC but a pro-inflammatory one in HCAEC. Our study highlights that, although they are both arterial EC, female HAoEC and HCAEC are distinguishable with regard to the role of EGFR and its involvement in inflammation regulation, what may be relevant for vascular maintenance but also the pathogenesis of endothelial dysfunction.
Collapse
Affiliation(s)
- Virginie Dubourg
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany.
| | - Gerald Schwerdt
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany
| | - Michael Kopf
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany
| | - Ralf A Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112, Halle, Germany
| |
Collapse
|
3
|
Stangl V, Lorenz M. Why you should pay more attention to your cells' sex. J Physiol 2023; 601:5581-5584. [PMID: 37963009 DOI: 10.1113/jp284819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Affiliation(s)
- Verena Stangl
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Mario Lorenz
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
4
|
Zhou C, Freel C, Mills O, Yang XR, Yan Q, Zheng J. MicroRNA-29 differentially mediates preeclampsia-dysregulated cellular responses to cytokines in female and male fetal endothelial cells. J Physiol 2023; 601:3631-3645. [PMID: 37401732 PMCID: PMC10807859 DOI: 10.1113/jp284746] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
Preeclampsia (PE) differentially impairs female and male fetal endothelial cell function, which is associated with an increased risk of adult-onset cardiovascular disorders in children born to mothers with PE. However, the underlying mechanisms are poorly defined. We hypothesize that dysregulation of microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in PE disturbs gene expression and cellular responses to cytokines in fetal endothelial cells in a fetal sex-dependent manner. RT-qPCR analysis of miR-29a/c-3p was performed on female and male unpassaged (P0) human umbilical vein endothelial cells (HUVECs) from normotensive (NT) pregnancies and PE. Bioinformatic analysis of an RNA-seq dataset was performed to identify PE-dysregulated miR-29a/c-3p target genes in female and male P0-HUVECs. Gain- and loss-of-function assays were conducted to determine the effects of miR-29a/c-3p on endothelial monolayer integrity and proliferation in response to transforming growth factor-β1 (TGFβ1) and tumour necrosis factor-α (TNFα) in NT and PE HUVECs at passage 1. We observed that PE downregulated miR-29a/c-3p in male and female P0-HUVECs. PE dysregulated significantly more miR-29a/c-3p target genes in female vs. male P0-HUVECs. Many of these PE-differentially dysregulated miR-29a/c-3p target genes are associated with critical cardiovascular diseases and endothelial function. We further demonstrated that miR-29a/c-3p knockdown specifically recovered the PE-abolished TGFβ1-induced strengthening of endothelial monolayer integrity in female HUVECs, while miR-29a/c-3p overexpression specifically enhanced the TNFα-promoted cell proliferation in male PE HUVECs. In conclusion, PE downregulates miR-29a/c-3p expression and differentially dysregulates miR-29a/c-3p target genes associated with cardiovascular diseases and endothelial function in female and male fetal endothelial cells, possibly contributing to the fetal sex-specific endothelial dysfunction observed in PE. KEY POINTS: Preeclampsia differentially impairs female and male fetal endothelial cell function in responses to cytokines. Pro-inflammatory cytokines are elevated in maternal circulation during pregnancy in preeclampsia. MicroRNAs are critical regulators of endothelial cell function during pregnancy. We have previously reported that preeclampsia downregulated microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in primary fetal endothelial cells. However, it is unknown if PE differentially dysregulates the expression of miR-29a/c-3p in female and male fetal endothelial cells. We show that preeclampsia downregulates miR-29a/c-3p in male and female HUVECs and preeclampsia dysregulates cardiovascular disease- and endothelial function-associated miR-29a/c-3p target genes in HUVECs in a fetal sex-specific manner. MiR-29a/c-3p differentially mediate cell responses to cytokines in female and male fetal endothelial cells from preeclampsia. We have revealed fetal sex-specific dysregulation of miR-29a/c-3p target genes in fetal endothelial cells from preeclampsia. This differential dysregulation may contribute to fetal sex-specific endothelial dysfunction in offspring born to preeclamptic mothers.
Collapse
Affiliation(s)
- Chi Zhou
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
- Department of Obstetrics and Gynecology, the University of Arizona, Tucson, AZ, United States
| | - Colman Freel
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Current Institution: University of Nebraska Medical Center, Omaha, NE, United States
| | - Olivia Mills
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Xin-Ran Yang
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Qin Yan
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
5
|
Abstract
Although sex differences have been noted in cellular function and behavior, therapy efficacy, and disease incidence and outcomes, the adoption of sex as a biological variable in tissue engineering and regenerative medicine remains limited. Furthering the development of personalized, precision medicine requires considering biological sex at the bench and in the clinic. This review provides the basis for considering biological sex when designing tissue-engineered constructs and regenerative therapies by contextualizing sex as a biological variable within the tissue engineering triad of cells, matrices, and signals. To achieve equity in biological sex within medicine requires a cultural shift in science and engineering research, with active engagement by researchers, clinicians, companies, policymakers, and funding agencies.
Collapse
Affiliation(s)
- Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Christopher Ludtka
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Bryan D James
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA;
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA
| |
Collapse
|
6
|
Weber CM, Harris MN, Zic SM, Sangha GS, Arnold NS, Dluzen DF, Clyne AM. Angiotensin II Increases Oxidative Stress and Inflammation in Female, But Not Male, Endothelial Cells. Cell Mol Bioeng 2023; 16:127-141. [PMID: 37096068 PMCID: PMC10121986 DOI: 10.1007/s12195-023-00762-2] [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: 10/19/2022] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Introduction Women are at elevated risk for certain cardiovascular diseases, including pulmonary arterial hypertension, Alzheimer's disease, and vascular complications of diabetes. Angiotensin II (AngII), a circulating stress hormone, is elevated in cardiovascular disease; however, our knowledge of sex differences in the vascular effects of AngII are limited. We therefore analyzed sex differences in human endothelial cell response to AngII treatment. Methods Male and female endothelial cells were treated with AngII for 24 h and analyzed by RNA sequencing. We then used endothelial and mesenchymal markers, inflammation assays, and oxidative stress indicators to measure female and male endothelial cell functional changes in response to AngII. Results Our data show that female and male endothelial cells are transcriptomically distinct. Female endothelial cells treated with AngII had widespread gene expression changes related to inflammatory and oxidative stress pathways, while male endothelial cells had few gene expression changes. While both female and male endothelial cells maintained their endothelial phenotype with AngII treatment, female endothelial cells showed increased release of the inflammatory cytokine interleukin-6 and increased white blood cell adhesion following AngII treatment concurrent with a second inflammatory cytokine. Additionally, female endothelial cells had elevated reactive oxygen species production compared to male endothelial cells after AngII treatment, which may be partially due to nicotinamide adenine dinucleotide phosphate oxidase-2 (NOX2) escape from X-chromosome inactivation. Conclusions These data suggest that endothelial cells have sexually dimorphic responses to AngII, which could contribute to increased prevalence of some cardiovascular diseases in women. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00762-2.
Collapse
Affiliation(s)
- Callie M. Weber
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Mikayla N. Harris
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Sophia M. Zic
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Gurneet S. Sangha
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Nicole S. Arnold
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Douglas F. Dluzen
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| |
Collapse
|
7
|
Zhou C, Freel C, Mills O, Yang XR, Yan Q, Zheng J. MicroRNA-29 Differentially Mediates Preeclampsia-Dysregulated Cellular Responses to Cytokines in Female and Male Fetal Endothelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.532827. [PMID: 36993536 PMCID: PMC10055181 DOI: 10.1101/2023.03.17.532827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Introduction Preeclampsia (PE) differentially impairs female and male fetal endothelial cell function which is associated with the increased risks of adult-onset cardiovascular disorders in children born to mothers with PE. However, the underlying mechanisms are poorly defined. We hypothesize that dysregulation of microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in PE disturbs gene expression and cellular responses to cytokines in fetal endothelial cells in a fetal sex-dependent manner. Methods RT-qPCR analysis of miR-29a/c-3p was performed on female and male unpassaged (P0) human umbilical vein endothelial cells (HUVECs) from normotensive (NT) and PE pregnancies. Bioinformatic analysis of an RNAseq dataset was performed to identify PE-dysregulated miR-29a/c-3p target genes in female and male P0-HUVECs. Gain- and loss-of-function assays were conducted to determine the effects of miR-29a/c-3p on endothelial monolayer integrity and proliferation in response to TGFβ1 and TNFα in NT and PE HUVECs at passage 1. Results PE downregulated miR-29a/c-3p in male, but not female P0-HUVECs. PE dysregulated significantly more miR-29a/c-3p target genes in female vs. male P0-HUVECs. Many of these PE-differentially dysregulated miR-29a/c-3p target genes are associated with critical cardiovascular diseases and endothelial functions. We further demonstrated that miR-29a/c-3p knockdown specifically recovered the PE-abolished TGFβ1-induced strengthening of endothelial monolayer integrity in female HUVECs, while miR-29a/c-3p overexpression specifically enhanced the TNFα-promoted cell proliferation in male PE HUVECs. Conclusions PE differentially dysregulates miR-29a/c-3p and their target genes associated with cardiovascular diseases- and endothelial function in female and male fetal endothelial cells, possibly contributing to the fetal sex-specific endothelial dysfunction observed in PE.
Collapse
|
8
|
Hayward-Piatkovskyi B, Gonyea CR, Pyle SC, Lingappan K, Gleghorn JP. Sex-related external factors influence pulmonary vascular angiogenesis in a sex-dependent manner. Am J Physiol Heart Circ Physiol 2023; 324:H26-H32. [PMID: 36367696 PMCID: PMC9762957 DOI: 10.1152/ajpheart.00552.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a disease with a significant sexual dimorphism where males have a disadvantage compared with their female counterparts. Although mechanisms behind this sexual dimorphism are poorly understood, sex differences in angiogenesis have been identified as one possible source of the male disadvantage in BPD. Pulmonary angiogenesis was assessed in vitro using a bead sprouting assay with pooled male or female human pulmonary microvascular endothelial cells (HPMECs, 18-19 wk gestation, canalicular stage of human lung development) in standard (sex-hormone containing) and hormone-stripped medium. We identified sex-specific phenotypes in angiogenesis where male HPMECs produce fewer but longer sprouts compared with female HPMECs. The presence of sex hormones from standard culture medium modifies the male HPMEC phenotype with shorter and fewer sprouts but does not influence the female phenotype. Using a conditioned medium model, we further characterized the influence of the sex-specific secretome. Male and female HPMECs secrete factors that increase the maximum length of sprouts in female, but not male HPMECs. The presence of sex hormones abolishes this response. The male HPMEC secretome inhibits angiogenic sprouting in male HPMECs in the absence of sex hormones. Taken together, these results demonstrate that the pulmonary endothelial cell phenotypes are influenced by sex hormones and sex-specific secreted factors in a sex-dependent manner.NEW & NOTEWORTHY We identified a sex-specific phenotype wherein male HPMECs produce fewer but longer sprouts than females. Surprisingly, the presence of sex hormones only modifies the male phenotype, resulting in shorter and even fewer sprouts. Furthermore, we found the sex-specific secretome has a sex-dependent influence on angiogenesis that is also sex-hormone sensitive. These new and surprising findings point to the unappreciated role of sex and sex-related exogenous factors in early developmental angiogenesis.
Collapse
Affiliation(s)
| | - Cailin R Gonyea
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Sienna C Pyle
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Krithika Lingappan
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason P Gleghorn
- Department of Biological Sciences, University of Delaware, Newark, Delaware
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| |
Collapse
|
9
|
Rudnicki M, Pislaru A, Rezvan O, Rullman E, Fawzy A, Nwadozi E, Roudier E, Gustafsson T, Haas TL. Transcriptomic profiling reveals sex-specific molecular signatures of adipose endothelial cells under obesogenic conditions. iScience 2022; 26:105811. [PMID: 36624843 PMCID: PMC9823135 DOI: 10.1016/j.isci.2022.105811] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/13/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Female mice display greater adipose angiogenesis and maintain healthier adipose tissue than do males upon high-fat diet feeding. Through transcriptome analysis of endothelial cells (EC) from the white adipose tissue of male and female mice high-fat-fed for 7 weeks, we found that adipose EC exhibited pronouncedly sex-distinct transcriptomes. Genes upregulated in female adipose EC were associated with proliferation, oxidative phosphorylation, and chromatin remodeling contrasting the dominant enrichment for genes related to inflammation and a senescence-associated secretory of male EC. Similar sex-biased phenotypes of adipose EC were detectable in a dataset of aged EC. The highly proliferative phenotype of female EC was observed also in culture conditions. In turn, male EC displayed greater inflammatory potential than female EC in culture, based on basal and tumor necrosis factor alpha-stimulated patterns of gene expression. Our study provides insights into molecular programs that distinguish male and female EC responses to pathophysiological conditions.
Collapse
Affiliation(s)
- Martina Rudnicki
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada,Corresponding author
| | | | - Omid Rezvan
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Eric Rullman
- Department Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Department Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Aly Fawzy
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Emmanuel Nwadozi
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Emilie Roudier
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Thomas Gustafsson
- Department Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Department Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Tara L. Haas
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada,Department of Biology, York University, Toronto, Canada,Corresponding author
| |
Collapse
|
10
|
Hyperuricemia and Endothelial Function: Is It a Simple Association or Do Gender Differences Play a Role in This Binomial? Biomedicines 2022; 10:biomedicines10123067. [PMID: 36551823 PMCID: PMC9775568 DOI: 10.3390/biomedicines10123067] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022] Open
Abstract
The endothelium plays a fundamental role in the biological processes that ensure physiological vessel integrity, synthesizing numerous substances that are capable of modulating the tone of vessels, inflammation and the immune system, and platelet function. Endothelial dysfunction refers to an anomaly that develops at the level of the tunica that lines the internal surface of arterial and venous vessels, or, more precisely, an alteration to normal endothelial function, which involves the loss of some structural and/or functional characteristics. Studies on sex differences in endothelial function are conflicting, with some showing an earlier decline in endothelial function in men compared to women, while others show a similar age of onset between the sexes. Since increased cardiovascular risk coincides with menopause, female hormones, particularly estrogen, are generally believed to be cardioprotective. Furthermore, it is often proposed that androgens are harmful. In truth, these relationships are more complex than one might think and are not just dependent on fluctuations in circulating hormones. An increase in serum uric acid is widely regarded as a possible risk factor for cardiovascular disease; however, its role in the occurrence of endothelial dysfunction has not yet been elucidated. Several studies in the literature have evaluated sex-related differences in the association between elevated uric acid levels and cardiovascular events, with conflicting results. The association between uric acid and cardiovascular disease is still controversial, and it is not yet clear how gender differences affect the serum concentration of these substances. This review was primarily aimed at clarifying the effects of uric acid at the level of the vascular endothelium and describing how it could theoretically cause damage to endothelial integrity. The second aim was to determine if there are gender differences in uric acid metabolism and how these differences interact with the vascular endothelium.
Collapse
|
11
|
Rasile M, Lauranzano E, Faggiani E, Ravanelli MM, Colombo FS, Mirabella F, Corradini I, Malosio ML, Borreca A, Focchi E, Pozzi D, Giorgino T, Barajon I, Matteoli M. Maternal immune activation leads to defective brain-blood vessels and intracerebral hemorrhages in male offspring. EMBO J 2022; 41:e111192. [PMID: 36314682 PMCID: PMC9713716 DOI: 10.15252/embj.2022111192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
Intracerebral hemorrhages are recognized risk factors for neurodevelopmental disorders and represent early biomarkers for cognitive dysfunction and mental disability, but the pathways leading to their occurrence are not well defined. We report that a single intrauterine exposure of the immunostimulant Poly I:C to pregnant mice at gestational day 9, which models a prenatal viral infection and the consequent maternal immune activation, induces the defective formation of brain vessels and causes intracerebral hemorrhagic events, specifically in male offspring. We demonstrate that maternal immune activation promotes the production of the TGF-β1 active form and the consequent enhancement of pSMAD1-5 in males' brain endothelial cells. TGF-β1, in combination with IL-1β, reduces the endothelial expression of CD146 and claudin-5, alters the endothelium-pericyte interplay resulting in low pericyte coverage, and increases hemorrhagic events in the adult offspring. By showing that exposure to Poly I:C at the beginning of fetal cerebral angiogenesis results in sex-specific alterations of brain vessels, we provide a mechanistic framework for the association between intragravidic infections and anomalies of the neural vasculature, which may contribute to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Marco Rasile
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | | | - Elisa Faggiani
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Margherita M Ravanelli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | | | - Filippo Mirabella
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Irene Corradini
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Maria L Malosio
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Antonella Borreca
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Elisa Focchi
- Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Davide Pozzi
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Toni Giorgino
- Institute of Biophysics (IBF‐CNR)National Research Council of ItalyMilanItaly
| | - Isabella Barajon
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Michela Matteoli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| |
Collapse
|
12
|
Dietrich E, Jomard A, Osto E. Crosstalk between high-density lipoproteins and endothelial cells in health and disease: Insights into sex-dependent modulation. Front Cardiovasc Med 2022; 9:989428. [PMID: 36304545 PMCID: PMC9594152 DOI: 10.3389/fcvm.2022.989428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death worldwide. Intense research in vascular biology has advanced our knowledge of molecular mechanisms of its onset and progression until complications; however, several aspects of the patho-physiology of atherosclerosis remain to be further elucidated. Endothelial cell homeostasis is fundamental to prevent atherosclerosis as the appearance of endothelial cell dysfunction is considered the first pro-atherosclerotic vascular modification. Physiologically, high density lipoproteins (HDLs) exert protective actions for vessels and in particular for ECs. Indeed, HDLs promote endothelial-dependent vasorelaxation, contribute to the regulation of vascular lipid metabolism, and have immune-modulatory, anti-inflammatory and anti-oxidative properties. Sex- and gender-dependent differences are increasingly recognized as important, although not fully elucidated, factors in cardiovascular health and disease patho-physiology. In this review, we highlight the importance of sex hormones and sex-specific gene expression in the regulation of HDL and EC cross-talk and their contribution to cardiovascular disease.
Collapse
Affiliation(s)
- Elisa Dietrich
- Institute for Clinical Chemistry, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Anne Jomard
- Institute for Clinical Chemistry, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Elena Osto
- Institute for Clinical Chemistry, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Department of Cardiology, Heart Center, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Cox‐Flaherty K, Baird GL, Braza J, Guarino BD, Princiotto A, Ventetuolo CE, Harrington EO. Commercial human pulmonary artery endothelial cells have in-vitro behavior that varies by sex. Pulm Circ 2022; 12:e12165. [PMID: 36484057 PMCID: PMC9723258 DOI: 10.1002/pul2.12165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
It is unknown whether biological sex influences phenotypes of commercially available human pulmonary artery endothelial cells (HPAECs). Ten lots of commercial HPAECs were used (Lonza Biologics; PromoCell). Five (50%) were confirmed to be genotypically male (SRY+) and five (50%) were confirmed to be female (SRY-). Experiments were conducted between passages five and eight. HPAEC phenotype was confirmed with a panel of cell expression markers. Standard assays for proliferation, migration and tube formation were performed in triplicate with technical replicates, under three treatment conditions (EndoGRO; Sigma-Aldrich). Apoptosis was assessed by exposing cells treated with complete media or low serum media to hypoxic (1% oxygen) or normoxic (20% oxygen) conditions. Laboratory staff was blinded. The median (range) age of male and female donors from whom the HPAECs were derived was 58 (48-60) and 56 (33-67), respectively. Our results suggest decreased proliferation in genotypically female cells compared with male cells (p = 0.09). With increasing donor age, female cells were less proliferative and male cells were more proliferative (p = 0.001). Female cells were significantly more apoptotic than male cells by condition (p = 0.001). Female cells were significantly more migratory than male cells in complete media but less migratory than male cells under vascular endothelial growth factor enriched conditions (p = 0.001). There are subtle sex-based differences in the behavior of HPAECs that depend on donor sex and, less so, age. These differences may undermine rigor and reproducibility. Future studies should define whether biological sex is an important regulator of HPAEC function in health and disease.
Collapse
Affiliation(s)
- Katherine Cox‐Flaherty
- Departments of Medicine and Health ServicesPolicy and Practice, Brown UniversityProvidenceRhode IslandUSA
- Vascular Research Laboratory, Providence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
| | | | - Julie Braza
- Vascular Research Laboratory, Providence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
| | - Brianna D. Guarino
- Departments of Medicine and Health ServicesPolicy and Practice, Brown UniversityProvidenceRhode IslandUSA
- Vascular Research Laboratory, Providence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
| | - Amy Princiotto
- Vascular Research Laboratory, Providence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
| | - Corey E. Ventetuolo
- Departments of Medicine and Health ServicesPolicy and Practice, Brown UniversityProvidenceRhode IslandUSA
- Health Services, Policy and PracticeBrown UniversityProvidenceRhode IslandUSA
| | - Elizabeth O. Harrington
- Departments of Medicine and Health ServicesPolicy and Practice, Brown UniversityProvidenceRhode IslandUSA
- Vascular Research Laboratory, Providence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
| |
Collapse
|
14
|
Mendez PL, Obendorf L, Jatzlau J, Burdzinski W, Reichenbach M, Nageswaran V, Haghikia A, Stangl V, Hiepen C, Knaus P. Atheroprone fluid shear stress-regulated ALK1-Endoglin-SMAD signaling originates from early endosomes. BMC Biol 2022; 20:210. [PMID: 36171573 PMCID: PMC9520843 DOI: 10.1186/s12915-022-01396-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fluid shear stress enhances endothelial SMAD1/5 signaling via the BMP9-bound ALK1 receptor complex supported by the co-receptor Endoglin. While moderate SMAD1/5 activation is required to maintain endothelial quiescence, excessive SMAD1/5 signaling promotes endothelial dysfunction. Increased BMP signaling participates in endothelial-to-mesenchymal transition and inflammation culminating in vascular diseases such as atherosclerosis. While the function of Endoglin has so far been described under picomolar concentrations of BMP9 and short-term shear application, we investigated Endoglin under physiological BMP9 and long-term pathophysiological shear conditions. RESULTS We report here that knock-down of Endoglin leads to exacerbated SMAD1/5 phosphorylation and atheroprone gene expression profile in HUVECs sheared for 24 h. Making use of the ligand-trap ALK1-Fc, we furthermore show that this increase is dependent on BMP9/10. Mechanistically, we reveal that long-term exposure of ECs to low laminar shear stress leads to enhanced Endoglin expression and endocytosis of Endoglin in Caveolin-1-positive early endosomes. In these endosomes, we could localize the ALK1-Endoglin complex, labeled BMP9 as well as SMAD1, highlighting Caveolin-1 vesicles as a SMAD signaling compartment in cells exposed to low atheroprone laminar shear stress. CONCLUSIONS We identified Endoglin to be essential in preventing excessive activation of SMAD1/5 under physiological flow conditions and Caveolin-1-positive early endosomes as a new flow-regulated signaling compartment for BMP9-ALK1-Endoglin signaling axis in atheroprone flow conditions.
Collapse
Affiliation(s)
- Paul-Lennard Mendez
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- International Max-Planck Research School for Biology and Computation, Berlin, Germany
| | - Leon Obendorf
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Wiktor Burdzinski
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin School for Regenerative Therapies, Berlin, Germany
| | - Maria Reichenbach
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Vanasa Nageswaran
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Verena Stangl
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Faculty of Engineering and Natural Sciences, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany.
- International Max-Planck Research School for Biology and Computation, Berlin, Germany.
- Berlin School for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
15
|
Lorenz M, Witt E, Völker U, Stangl K, Stangl V, Hammer E. Serum starvation induces sexual dimorphisms in secreted proteins of human umbilical vein endothelial cells (HUVECs) from twin pairs. Proteomics 2022; 22:e2100168. [PMID: 35357760 DOI: 10.1002/pmic.202100168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/06/2022]
Abstract
There is growing evidence for sex and gender differences in the clinical manifestation and outcomes of human diseases. Human primary endothelial cells represent a useful cardiovascular model to study sexual dimorphisms at the cellular level. Here, we analyzed sexual dimorphisms of the secretome after serum starvation using HUVECs from twin pairs of the opposite sex to minimize the impact of varying genetic background. HUVECs were starved for 5 and 16 h, respectively, and proteins of the cell culture supernatants were analyzed by tandem mass spectrometry. Altogether, 960 extracellular proteins were identified of which 683 were amendable to stringent quantification. Significant alterations were observed for 324 proteins between long-term and short-term starvation and the majority were similar in both sexes. Only 5 proteins showed significant sex-specific regulation between long- versus short-term starvation. Furthermore, 19 unique proteins with significant sexual dimorphisms at the same time points of serum starvation were observed. A larger number of proteins, e.g. tissue factor inhibitor 2 (TFPI2), displayed higher levels in the supernatants of females compared to male cells after long term serum starvation that might point to higher adaptation capacity of female cells. The overall results demonstrate that male and female cells differ in their secretome. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mario Lorenz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Eric Witt
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| | - Karl Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Verena Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| |
Collapse
|
16
|
Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm. Biomolecules 2022; 12:biom12040509. [PMID: 35454098 PMCID: PMC9030795 DOI: 10.3390/biom12040509] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/27/2022] [Indexed: 12/25/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), defined as a focal dilation of the abdominal aorta beyond 50% of its normal diameter, is a common and potentially life-threatening vascular disease. The molecular and cellular mechanisms underlying AAA pathogenesis remain unclear. Healthy endothelial cells (ECs) play a critical role in maintaining vascular homeostasis by regulating vascular tone and maintaining an anti-inflammatory, anti-thrombotic local environment. Increasing evidence indicates that endothelial dysfunction is an early pathologic event in AAA formation, contributing to both oxidative stress and inflammation in the degenerating arterial wall. Recent studies utilizing single-cell RNA sequencing revealed heterogeneous EC sub-populations, as determined by their transcriptional profiles, in aortic aneurysm tissue. This review summarizes recent findings, including clinical evidence of endothelial dysfunction in AAA, the impact of biomechanical stress on EC in AAA, the role of endothelial nitric oxide synthase (eNOS) uncoupling in AAA, and EC heterogeneity in AAA. These studies help to improve our understanding of AAA pathogenesis and ultimately may lead to the generation of EC-targeted therapeutics to treat or prevent this deadly disease.
Collapse
|
17
|
Exposome and foetoplacental vascular dysfunction in gestational diabetes mellitus. Mol Aspects Med 2021; 87:101019. [PMID: 34483008 DOI: 10.1016/j.mam.2021.101019] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022]
Abstract
A balanced communication between the mother, placenta and foetus is crucial to reach a successful pregnancy. Several windows of exposure to environmental toxins are present during pregnancy. When the women metabolic status is affected by a disease or environmental toxin, the foetus is impacted and may result in altered development and growth. Gestational diabetes mellitus (GDM) is a disease of pregnancy characterised by abnormal glucose metabolism affecting the mother and foetus. This disease of pregnancy associates with postnatal consequences for the child and the mother. The whole endogenous and exogenous environmental factors is defined as the exposome. Endogenous insults conform to the endo-exposome, and disruptors contained in the immediate environment are the ecto-exposome. Some components of the endo-exposome, such as Selenium, vitamins D and B12, adenosine, and a high-fat diet, and ecto-exposome, such as the heavy metals Arsenic, Mercury, Lead and Copper, and per- and polyfluoroakyl substances, result in adverse pregnancies, including an elevated risk of GDM or gestational diabesity. The impact of the exposome on the human placenta's vascular physiology and function in GDM and gestational diabesity is reviewed.
Collapse
|
18
|
James BD, Allen JB. Sex-Specific Response to Combinations of Shear Stress and Substrate Stiffness by Endothelial Cells In Vitro. Adv Healthc Mater 2021; 10:e2100735. [PMID: 34142471 PMCID: PMC8458248 DOI: 10.1002/adhm.202100735] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 12/25/2022]
Abstract
By using a full factorial design of experiment, the combinatorial effects of biological sex, shear stress, and substrate stiffness on human umbilical vein endothelial cell (HUVEC) spreading and Yes-associated protein 1 (YAP1) activity are able to be efficiently evaluated. Within the range of shear stress (0.5-1.5 Pa) and substrate stiffness (10-100 kPa), male HUVECs are smaller than female HUVECs. Only with sufficient mechanical stimulation do they spread to a similar size. More importantly, YAP1 nuclear localization in female HUVECs is invariant to mechanical stimulation within the range of tested conditions whereas for male HUVECs it increases nonlinearly with increasing shear stress and substrate stiffness. The sex-specific response of HUVECs to combinations of shear stress and substrate stiffness reinforces the need to include sex as a biological variable and multiple mechanical stimuli in experiments, informs the design of precision biomaterials, and offers insight for understanding cardiovascular disease sexual dimorphisms. Moreover, here it is illustrated that different complex mechanical microenvironments can lead to sex-specific phenotypes and sex invariant phenotypes in cultured endothelial cells.
Collapse
Affiliation(s)
- Bryan D James
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| |
Collapse
|
19
|
Tan CMJ, Lewandowski AJ, Williamson W, Huckstep OJ, Yu GZ, Fischer R, Simon JN, Alsharqi M, Mohamed A, Leeson P, Bertagnolli M. Proteomic Signature of Dysfunctional Circulating Endothelial Colony-Forming Cells of Young Adults. J Am Heart Assoc 2021; 10:e021119. [PMID: 34275329 PMCID: PMC8475699 DOI: 10.1161/jaha.121.021119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Background A subpopulation of endothelial progenitor cells called endothelial colony-forming cells (ECFCs) may offer a platform for cellular assessment in clinical studies because of their remarkable angiogenic and expansion potentials in vitro. Despite endothelial cell function being influenced by cardiovascular risk factors, no studies have yet provided a comprehensive proteomic profile to distinguish functional (ie, more angiogenic and expansive cells) versus dysfunctional circulating ECFCs of young adults. The aim of this study was to provide a detailed proteomic comparison between functional and dysfunctional ECFCs. Methods and Results Peripheral blood ECFCs were isolated from 11 subjects (45% men, aged 27±5 years) using Ficoll density gradient centrifugation. ECFCs expressed endothelial and progenitor surface markers and displayed cobblestone-patterned morphology with clonal and angiogenic capacities in vitro. ECFCs were deemed dysfunctional if <1 closed tube formed during the in vitro tube formation assay and proliferation rate was <20%. Hierarchical functional clustering revealed distinct ECFC proteomic signatures between functional and dysfunctional ECFCs with changes in cellular mechanisms involved in exocytosis, vesicle transport, extracellular matrix organization, cell metabolism, and apoptosis. Targeted antiangiogenic proteins in dysfunctional ECFCs included SPARC (secreted protein acidic and rich in cysteine), CD36 (cluster of differentiation 36), LUM (lumican), and PTX3 (pentraxin-related protein PYX3). Conclusions Circulating ECFCs with impaired angiogenesis and expansion capacities have a distinct proteomic profile and significant phenotype changes compared with highly angiogenic endothelial cells. Impaired angiogenesis in dysfunctional ECFCs may underlie the link between endothelial dysfunction and cardiovascular disease risks in young adults.
Collapse
Affiliation(s)
- Cheryl M. J. Tan
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Adam J. Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Wilby Williamson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Odaro J. Huckstep
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of BiologyUnited States Air Force AcademyColorado SpringsCOUSA
| | - Grace Z. Yu
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Roman Fischer
- Target Discovery Institute (TDI) Mass Spectrometry Laboratory, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Jillian N. Simon
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Maryam Alsharqi
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Cardiac TechnologyImam Abdulrahman Bin Faisal UniversityDammamSaudi Arabia
| | - Afifah Mohamed
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Diagnostic Imaging & Applied Health Sciences, Faculty of Health SciencesUniversiti Kebangsaan MalaysiaKuala LumpurMalaysia
| | - Paul Leeson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Mariane Bertagnolli
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Montreal Hospital Sacré‐Cœur Research CentreCentre Intégré Universitaire de Santé et de Services Sociaux du Nord‐de‐l'Île‐de‐MontréalMontréalQCCanada
- School of Physical and Occupational Therapy, Faculty of MedicineMcGill UniversityMontréalQCCanada
| |
Collapse
|
20
|
Keuper M, Jastroch M. The good and the BAT of metabolic sex differences in thermogenic human adipose tissue. Mol Cell Endocrinol 2021; 533:111337. [PMID: 34062167 DOI: 10.1016/j.mce.2021.111337] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022]
Abstract
Thermogenic adipose tissue, which comprises classical brown and beige adipose tissue, has the ability to improve systemic metabolism. Its identification in adult humans has fostered extensive investigations on the therapeutic value to counteract obesity and metabolic disorders. Sex and gender differences of human thermogenic adipose tissue, however, are still understudied despite their importance for personalized treatment options. Here, we review studies reporting human sex differences of thermogenic adipose tissue and related potential improvements of systemic energy metabolism. An increasing body of evidence suggests higher prevalence, mass and activity of thermogenic adipose tissue in women, but the consequences for metabolic disease progression and mechanisms are largely unknown. Therefore, we also discuss observations on sex-specific adipose metabolism in experimental mouse and rat studies that may assist to establish molecular mechanisms and instruct future investigations in humans.
Collapse
Affiliation(s)
- Michaela Keuper
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden.
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| |
Collapse
|
21
|
Erfinanda L, Ravindran K, Kohse F, Gallo K, Preissner R, Walther T, Kuebler WM. Oestrogen-mediated upregulation of the Mas receptor contributes to sex differences in acute lung injury and lung vascular barrier regulation. Eur Respir J 2021; 57:13993003.00921-2020. [PMID: 32764118 DOI: 10.1183/13993003.00921-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/30/2020] [Indexed: 11/05/2022]
Abstract
Epidemiological data from the SARS-CoV-2 outbreak suggest sex differences in mortality and vulnerability; however, sex-dependent incidence of acute respiratory distress syndrome (ARDS) remains controversial and the sex-dependent mechanisms of endothelial barrier regulation are unknown. In premenopausal women, increased signalling of angiotensin (Ang)(1-7) via the Mas receptor has been linked to lower cardiovascular risk. Since stimulation of the Ang(1-7)/Mas axis protects the endothelial barrier in acute lung injury (ALI), we hypothesised that increased Ang(1-7)/Mas signalling may protect females over males in ALI/ARDS.Clinical data were collected from Charité inpatients (Berlin) and sex differences in ALI were assessed in wild-type (WT) and Mas-receptor deficient (Mas-/- ) mice. Endothelial permeability was assessed as weight change in isolated lungs and as transendothelial electrical resistance (TEER) in vitroIn 734 090 Charité inpatients (2005-2016), ARDS had a higher incidence in men as compared to women. In murine ALI, male WT mice had more lung oedema, protein leaks and histological evidence of injury than female WT mice. Lung weight change in response to platelet-activating factor (PAF) was more pronounced in male WT and female Mas-/- mice than in female WT mice, whereas Mas-receptor expression was higher in female WT lungs. Ovariectomy attenuated protection in female WT mice and reduced Mas-receptor expression. Oestrogen increased Mas-receptor expression and attenuated endothelial leakage in response to thrombin in vitro This effect was alleviated by Mas-receptor blockade.Improved lung endothelial barrier function protects female mice from ALI-induced lung oedema. This effect is partially mediated via enhanced Ang(1-7)/Mas signalling as a result of oestrogen-dependent Mas expression.
Collapse
Affiliation(s)
- Lasti Erfinanda
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Krishnan Ravindran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Franziska Kohse
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany.,Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Kathleen Gallo
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Robert Preissner
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Thomas Walther
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany .,Dept of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland.,Shared senior authorship
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Dept of Physiology and Dept of Surgery, University of Toronto, Toronto, ON, Canada.,Shared senior authorship
| |
Collapse
|
22
|
Human Umbilical Cord: Information Mine in Sex-Specific Medicine. Life (Basel) 2021; 11:life11010052. [PMID: 33451112 PMCID: PMC7828611 DOI: 10.3390/life11010052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Biological differences between sexes should be considered in all stages of research, as sexual dimorphism starts in utero leading to sex-specific fetal programming. In numerous biomedical fields, there is still a lack of stratification by sex despite primary cultured cells retaining memory of the sex and of the donor. The sex of donors in biological research must be known because variations in cells and cellular components can be used as endpoints, biomarkers and/or targets of pharmacological studies. This selective review focuses on the current findings regarding sex differences observed in the umbilical cord, a widely used source of research samples, both in the blood and in the circulating cells, as well as in the different cellular models obtainable from it. Moreover, an overview on sex differences in fetal programming is reported. As it emerges that the sex variable is still often forgotten in experimental models, we suggest that it should be mandatory to adopt sex-oriented research, because only awareness of these issues can lead to innovative research.
Collapse
|
23
|
Cattaneo MG, Banfi C, Brioschi M, Lattuada D, Vicentini LM. Sex-dependent differences in the secretome of human endothelial cells. Biol Sex Differ 2021; 12:7. [PMID: 33413676 PMCID: PMC7791663 DOI: 10.1186/s13293-020-00350-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cellular sex has rarely been considered as a biological variable in preclinical research, even when the pathogenesis of diseases with predictable sex differences is studied. In this perspective, proteomics, and “omics” approaches in general, can provide powerful tools to obtain comprehensive cellular maps, thus favoring the discovery of still unknown sex-biased physio-pathological mechanisms. Methods We performed proteomic and Gene Ontology (GO) analyses of the secretome from human serum-deprived male and female endothelial cells (ECs) followed by ELISA validation. Apoptosis was detected by FACS and Western blot techniques and efferocytosis through the ability of the macrophage cell line RAW 264.7 to engulf apoptotic ECs. PTX3 mRNA levels were measured by RT-qPCR. Results Proteomic and GO analyses of the secretome from starved human male and female ECs demonstrated a significant enrichment in proteins related to cellular responses to stress and to the regulation of apoptosis in the secretome of male ECs. Accordingly, a higher percentage of male ECs underwent apoptosis in response to serum deprivation in comparison with female ECs. Among the secreted proteins, we reliably found higher levels of PTX3 in the male EC secretome. The silencing of PTX3 suggested that male ECs were dependent on its expression to properly carry out the efferocytotic process. At variance, female EC efferocytosis seemed to be independent on PTX3 expression. Conclusions Our results demonstrated that serum-starved male and female ECs possess different secretory phenotypes that might take part in the sex-biased response to cellular stress. We identified PTX3 as a crucial player in the male-specific endothelial response to an apoptotic trigger. This novel and sex-related role for secreted proteins, and mainly for PTX3, may open the way to the discovery of still unknown sex-specific mechanisms and pharmacological targets for the prevention and treatment of endothelial dysfunction at the onset of atherosclerosis and cardiovascular disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-020-00350-3.
Collapse
Affiliation(s)
- Maria Grazia Cattaneo
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy.
| | | | | | - Donatella Lattuada
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy
| | - Lucia M Vicentini
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy
| |
Collapse
|
24
|
James BD, Guerin P, Allen JB. Let's Talk About Sex-Biological Sex Is Underreported in Biomaterial Studies. Adv Healthc Mater 2021; 10:e2001034. [PMID: 33043626 PMCID: PMC7791002 DOI: 10.1002/adhm.202001034] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/26/2020] [Indexed: 12/13/2022]
Abstract
Precision medicine aims to better individualize healthcare. It requires that biomaterials be designed for the physiological characteristics of a specific patient. To make this a reality, biomaterials research and development must address differences of biological sex. More specifically, biomaterials should be designed with properties optimized and appropriate for male and female patients. In analyzing research articles from seven prominent biomaterials journals, sex as a biological variable is missing from an overwhelming majority of in vitro biomaterial studies. From the survey, the reporting of the sex of primary cell cultures happened only 10.3% of the time. Contributing to this trend is that commercial vendors bias cell lines toward one sex or another by not disclosing information of cell line sex at the time of purchase; researchers do not communicate this pertinent information in published studies; and many journal policies have little to no requirements for reporting cell line characteristics. Omitting this valuable information leads to a gap in the understanding of sex-specific cell-biomaterial interactions and it creates a bias in research findings towards one sex or another. To curb this concerning trend and make precision biomaterials a reality will require the biomaterials field to "talk about sex" by reporting cell sex more broadly.
Collapse
Affiliation(s)
- Bryan D James
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Paxton Guerin
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| |
Collapse
|
25
|
Boscaro C, Trenti A, Baggio C, Scapin C, Trevisi L, Cignarella A, Bolego C. Sex Differences in the Pro-Angiogenic Response of Human Endothelial Cells: Focus on PFKFB3 and FAK Activation. Front Pharmacol 2020; 11:587221. [PMID: 33390959 PMCID: PMC7773665 DOI: 10.3389/fphar.2020.587221] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/06/2020] [Indexed: 01/14/2023] Open
Abstract
Female hormones and sex-specific factors are established determinants of endothelial function, yet their relative contribution to human endothelium phenotypes has not been defined. Using human umbilical vein endothelial cells (HUVECs) genotyped by donor's sex, we investigated the influence of sex and estrogenic agents on the main steps of the angiogenic process and on key proteins governing HUVEC metabolism and migratory properties. HUVECs from female donors (fHUVECs) showed increased viability (p < 0.01) and growth rate (p < 0.01) compared with those from males (mHUVECs). Despite higher levels of G-protein coupled estrogen receptor (GPER) in fHUVECs (p < 0.001), treatment with 17β-estradiol (E2) and the selective GPER agonist G1 (both 1-100 nM) did not affect HUVEC viability. Migration and tubularization in vitro under physiological conditions were higher in fHUVECs than in mHUVECs (p < 0.05). E2 treatment (1-100 nM) upregulated the glycolytic activator PFKFB3 with higher potency in fHUVECs than in mHUVECs, despite comparable baseline levels. Moreover, Y576/577 phosphorylation of focal adhesion kinase (FAK) was markedly enhanced in fHUVECs (p < 0.001), despite comparable Src activation levels. While the PI3K inhibitor LY294002 (25 µM) inhibited HUVEC migration (p < 0.05), Akt phosphorylation levels in fHUVECs and mHUVECs were comparable. Finally, digitoxin treatment, which inhibits Y576/577 FAK phosphorylation, abolished sexual dimorphism in HUVEC migration. These findings unravel complementary modulation of HUVEC functional phenotypes and signaling molecules involved in angiogenesis by hormone microenvironment and sex-specific factors, and highlight the need for sex-oriented pharmacological targeting of endothelial function.
Collapse
Affiliation(s)
- Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Chiara Baggio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Chiara Scapin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
26
|
Salsoso R, Mate A, Toledo F, Vázquez CM, Sobrevia L. Insulin requires A 2B adenosine receptors to modulate the L-arginine/nitric oxide signalling in the human fetoplacental vascular endothelium from late-onset preeclampsia. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165993. [PMID: 33096224 DOI: 10.1016/j.bbadis.2020.165993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
Late-onset preeclampsia (LOPE) associates with reduced umbilical vein reactivity and endothelial nitric oxide synthase (eNOS) activity but increased human cationic amino acid (hCAT-1)-mediated L-arginine transport involving A2A adenosine receptor in the fetoplacental unit. This study addresses the A2B adenosine receptor (A2BAR)-mediated response to insulin in the fetoplacental vasculature from LOPE. Umbilical veins and HUVECs were obtained from women with normal (n = 37) or LOPE (n = 35) pregnancies. Umbilical vein rings reactivity to insulin was assayed in the absence or presence of adenosine and MRS-1754 (A2BAR antagonist) in a wire myograph. HUVECs were exposed to insulin, MRS-1754, BAY60-6583 (A2BAR agonist), NECA (general adenosine receptors agonist) or NG-nitro-L-arginine methyl ester (NOS inhibitor). A2BAR, hCAT-1, total and phosphorylated eNOS, Akt and p44/42mapk protein abundance were determined by Western blotting. Insulin receptors A (IR-A) and B (IR-B), eNOS and hCAT-1 mRNA were determined by qPCR. Firefly/Renilla luciferase assay was used to determine -1606 bp SLC7A1 (hCAT-1) promoter activity. L-Citrulline content was measured by HPLC, L-[3H]citrulline formation from L-[3H]arginine by the Citrulline assay, and intracellular cGMP by radioimmunoassay. LOPE-reduced dilation of vein rings to insulin was restored by MRS-1754. HUVECs from LOPE showed higher A2BAR, hCAT-1, and IR-A expression, Akt and p44/42mapk activation, and lower NOS activity. MRS-1754 reversed the LOPE effect on A2BAR, hCAT-1, Akt, and eNOS inhibitory phosphorylation. Insulin reversed the LOPE effect on A2BAR, IR-A and eNOS, but increased hCAT-1-mediated transport. Thus, LOPE alters endothelial function, causing an imbalance in the L-arginine/NO signalling pathway to reduce the umbilical vein dilation to insulin requiring A2BAR activation in HUVECs.
Collapse
Affiliation(s)
- Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain
| | - Alfonso Mate
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Carmen M Vázquez
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, Herston, QLD, 4029, Australia.
| |
Collapse
|
27
|
Advantages in Wound Healing Process in Female Mice Require Upregulation A 2A-Mediated Angiogenesis under the Stimulation of 17β-Estradiol. Int J Mol Sci 2020; 21:ijms21197145. [PMID: 32998232 PMCID: PMC7583763 DOI: 10.3390/ijms21197145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/12/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Estrogenic steroids and adenosine A2A receptors promote the wound healing and angiogenesis processes. However, so far, it is unclear whether estrogen may regulate the expression and pro-angiogenic activity of A2A receptors. Using in vivo analyses, we showed that female wild type (WT) mice have a more rapid wound healing process than female or male A2A-deficient mice (A2AKO) mice. We also found that pulmonary endothelial cells (mPEC) isolated from female WT mice showed higher expression of A2A receptor than mPEC from male WT mice. mPEC from female WT mice were more sensitive to A2A-mediated pro-angiogenic response, suggesting an ER and A2A crosstalk, which was confirmed using cells isolated from A2AKO. In those female cells, 17β-estradiol potentiated A2A-mediated cell proliferation, an effect that was inhibited by selective antagonists of estrogen receptors (ER), ERα, and ERβ. Therefore, estrogen regulates the expression and/or pro-angiogenic activity of A2A adenosine receptors, likely involving activation of ERα and ERβ receptors. Sexual dimorphism in wound healing observed in the A2AKO mice process reinforces the functional crosstalk between ER and A2A receptors.
Collapse
|
28
|
The protective role of estrogen on endothelial and glycocalyx barriers after shock conditions: A microfluidic study. Surgery 2020; 169:678-685. [PMID: 32988619 DOI: 10.1016/j.surg.2020.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/22/2020] [Accepted: 08/04/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Sexual dimorphism has been demonstrated after major trauma and hemorrhage shock with protective effects related to female sex or estrogen. Traumatic endotheliopathy is an important component of trauma-induced coagulopathy. Components of endothelial barrier dysfunction include degradation of the endothelial glycocalyx and endothelial cellular injury. Estrogen modulates endothelial function via its membrane and cellular receptors. The effects of estrogen on the vascular endothelial barrier after trauma and hemorrhage shock are, however, unknown. This topic was studied in an in vitro model under flow conditions. METHODS Monolayers of human umbilical vein endothelial cells were established in microfluidic flow devices. After overnight perfusion, cell monolayers were subjected to normoxic or hypoxic perfusion and then treated with either estrogen (as estradiol), testosterone (as dihydrotestosterone), or media alone. Endothelial activation/injury was indexed by soluble thrombomodulin and glycocalyx degradation by syndecan-1 and hyaluronic acid shedding as well as measurement of the thickness of the glycocalyx layer. The coagulation phenotype of the human umbilical vein endothelial cells was indexed by the relative values of the activities of tissue plasminogen activator and plasminogen activator inhibitor-1. Vascular endothelial growth factor was measured in cell culture supernatants using a solid-phase enzyme-linked immunosorbent assay. RESULTS Treatment with estrogen but not testosterone mitigated the adverse effect of shock on endothelial and glycocalyx barrier properties. Our biomimetic model suggests a beneficial effect of estrogen administration after trauma and hemorrhage shock on the glycocalyx and endothelial barriers. CONCLUSION Early estrogen treatment after trauma and hemorrhage shock may be a useful adjunct to mitigating the development of traumatic endotheliopathy.
Collapse
|
29
|
Hartman RJG, Kapteijn DMC, Haitjema S, Bekker MN, Mokry M, Pasterkamp G, Civelek M, den Ruijter HM. Intrinsic transcriptomic sex differences in human endothelial cells at birth and in adults are associated with coronary artery disease targets. Sci Rep 2020; 10:12367. [PMID: 32704153 PMCID: PMC7378217 DOI: 10.1038/s41598-020-69451-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Sex differences in endothelial cell (EC) biology may reflect intrinsic differences driven by chromosomes or sex steroid exposure and gender differences accumulated over life. We analysed EC gene expression data from boy-girl twins at birth and in non-twin adults to detect sex differences at different stages of life, and show that 14-25% of the EC transcriptome is sex-biased. By combining data from both stages of life, we identified sex differences that are present at birth and maintained throughout life, and those that are acquired over life. Promisingly, we found that genes that present with an acquired sex difference in ECs are more likely to be targets of sex steroids. Annotating both gene sets with data from multiple genome-wide association studies (GWAS) revealed that genes with an intrinsic sex difference in ECs are enriched for coronary artery disease GWAS hits. This study underscores the need for treating sex as a biological variable.
Collapse
Affiliation(s)
- Robin J G Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniek M C Kapteijn
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia Haitjema
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mireille N Bekker
- Department of Obstetrics and Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mete Civelek
- Center for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Division of Heart and Lungs, Department of Experimental Cardiology, University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, 3508GA, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Sexually dimorphic DNA-methylation in cardiometabolic health: A systematic review. Maturitas 2020; 135:6-26. [DOI: 10.1016/j.maturitas.2020.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
|
31
|
Kudze T, Ono S, Fereydooni A, Gonzalez L, Isaji T, Hu H, Yatsula B, Taniguchi R, Koizumi J, Nishibe T, Dardik A. Altered hemodynamics during arteriovenous fistula remodeling leads to reduced fistula patency in female mice. JVS Vasc Sci 2020; 1:42-56. [PMID: 32754721 PMCID: PMC7402599 DOI: 10.1016/j.jvssci.2020.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Objective The arteriovenous fistula (AVF) is the preferred method of dialysis access because of its proven superior long-term outcomes. However, women have lower rates of AVF patency and utilization than men. We used a novel mouse AVF model that recapitulates human AVF maturation to determine whether there are differences in AVF patency in female and male mice. Methods Aortocaval fistulas were created in female and male C57BL/6 mice (9-10 weeks). At days 0, 3, 7, and 21, infrarenal inferior vena cava (IVC) and aortic diameters and flow velocity were monitored by Doppler ultrasound and used to calculate the vessel diameter, blood flow, and shear stress. AVF were harvested, and expression of proteins was examined by proteomic analysis and immunofluorescence and of messenger RNA by quantitative polymerase chain reaction analysis. Results At baseline, female mice weighed less and had lower IVC velocity and smaller magnitudes of shear stress, but there was no significant difference in IVC diameter and thickness. After AVF creation, both female and male mice had similar IVC dilation and thickening with no significant differences in IVC wall thickness at day 21. However, female mice had diminished AVF patency by day 42 (25.7% vs 64.3%; P = .039). During fistula remodeling, female mice had lower IVC mean velocity and shear stress magnitude and increased spectral broadening (days 0-21). Messenger RNA and protein expression of Krüppel-like factor 2, endothelial nitric oxide synthase, and vascular cell adhesion molecule 1 was similar at baseline in female and male mice but increased in the AVF only in male mice but not in female mice (day 21). Proteomic analysis of female and male mice detected 56 proteins expressed at significantly higher levels in the IVC of female mice and 67 proteins expressed at significantly higher levels in the IVC of male mice (day 7); function-specific analysis showed that the IVC of male mice overexpressed proteins that belong to pathways implicated in the regulation of vascular function, thrombosis, response to flow, and vascular remodeling. Conclusions AVF in female mice have diminished patency, preceded by lower velocity, reduced magnitudes of shear stress, and less laminar flow during remodeling. There is also sex-specific differential expression of proteins involved in thrombosis, response to laminar flow, inflammation, and proliferation. These findings suggest that hemodynamic changes during fistula maturation may play an important role underlying the diminished rates of AVF utilization in women. Women have lower rates of arteriovenous fistula (AVF) utilization than men. Using a mouse AVF model that recapitulates human AVF maturation, we show that female mice have similar AVF remodeling but diminished patency. AVF remodeling in female mice is associated with reduced shear stress and laminar flow; lack of increased transcription and translation of several anti-inflammatory, antiproliferative, and laminar flow response proteins (endothelial nitric oxide synthase, Krüppel-like factor 2, and vascular cell adhesion molecule 1); and different patterns of expression of pathways that regulate thrombosis and venous remodeling. Identifying downstream targets involved in these mechanisms may improve AVF outcomes in female patients.
Collapse
Affiliation(s)
- Tambudzai Kudze
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Shun Ono
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Haidi Hu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Jun Koizumi
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara
| | - Toshiya Nishibe
- Department of Cardiovascular Surgery, Tokyo Medical University, Tokyo
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven
| |
Collapse
|
32
|
Use of Human Umbilical Vein Endothelial Cells (HUVEC) as a Model to Study Cardiovascular Disease: A Review. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10030938] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, and extensive research has been performed to understand this disease better, using various experimental models. The endothelium plays a crucial role in the development of CVD, since it is an interface between bloodstream components, such as monocytes and platelets, and other arterial wall components. Human umbilical vein endothelial cell (HUVEC) isolation from umbilical cord was first described in 1973. To date, this model is still widely used because of the high HUVEC isolation success rate, and because HUVEC are an excellent model to study a broad array of diseases, including cardiovascular and metabolic diseases. We here review the history of HUVEC isolation, the HUVEC model over time, HUVEC culture characteristics and conditions, advantages and disadvantages of this model and finally, its applications in the area of cardiovascular diseases.
Collapse
|
33
|
Green CE. Lung function and endothelial dysfunction: Is there a relationship without the presence of lung disease? Respirology 2020; 25:49-50. [DOI: 10.1111/resp.13573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Clara E. Green
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of Birmingham Birmingham UK
| |
Collapse
|
34
|
Lorenz M, Blaschke B, Benn A, Hammer E, Witt E, Kirwan J, Fritsche-Guenther R, Gloaguen Y, Bartsch C, Vietzke A, Kramer F, Kappert K, Brunner P, Nguyen HG, Dreger H, Stangl K, Knaus P, Stangl V. Sex-specific metabolic and functional differences in human umbilical vein endothelial cells from twin pairs. Atherosclerosis 2019; 291:99-106. [PMID: 31706078 DOI: 10.1016/j.atherosclerosis.2019.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/09/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Gonadal hormones are mainly thought to account for sex and gender differences in the incidence, clinical manifestation and therapy of many cardiovascular diseases. However, intrinsic sex differences at the cellular level are mostly overlooked. Here, we assessed sex-specific metabolic and functional differences between male and female human umbilical vein endothelial cells (HUVECs). METHODS Cellular metabolism was investigated by bioenergetic studies (Seahorse Analyser) and a metabolomic approach. Protein levels were determined by Western blots and proteome analysis. Vascular endothelial growth factor (VEGF)-stimulated cellular migration was assessed by gap closure. HUVECs from dizygotic twin pairs were used for most experiments. RESULTS No sex differences were observed in untreated cells. However, sexual dimorphisms appeared after stressing the cells by serum starvation and treatment with VEGF. Under both conditions, female cells had higher intracellular ATP and metabolite levels. A significant decline in ATP levels was observed in male cells after serum starvation. After VEGF, the ratio of glycolysis/mitochondrial respiration was higher in female cells and migration was more pronounced. CONCLUSIONS These results point to an increased stress tolerance of female cells. We therefore propose that female cells have an energetic advantage over male cells under conditions of diminished nutrient supply. A more favourable energy balance of female HUVECs after serum starvation and VEGF could potentially explain their stronger migratory capacity.
Collapse
Affiliation(s)
- Mario Lorenz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Benjamin Blaschke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Andreas Benn
- Institut für Chemie und Biochemie, Freie Universität Berlin, Germany; Berlin School of Integrative Oncology (DFG Graduate School 1093), Germany
| | - Elke Hammer
- Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Abteilung für Funktionelle Genomforschung, Universitätsmedizin Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Germany
| | - Eric Witt
- Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Abteilung für Funktionelle Genomforschung, Universitätsmedizin Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Germany
| | - Jennifer Kirwan
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany; Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany; Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Yoann Gloaguen
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany; Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany; Core Unit Bioinformatics, Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Cornelia Bartsch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Angelika Vietzke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Frederike Kramer
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Kai Kappert
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany; Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Patrizia Brunner
- Institut für Chemie und Biochemie, Freie Universität Berlin, Germany; Berlin School of Integrative Oncology (DFG Graduate School 1093), Germany
| | - Hoang Giang Nguyen
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Henryk Dreger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Karl Stangl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Petra Knaus
- Institut für Chemie und Biochemie, Freie Universität Berlin, Germany; Berlin School of Integrative Oncology (DFG Graduate School 1093), Germany
| | - Verena Stangl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany.
| |
Collapse
|
35
|
Reckelhoff JF, LaMarca B, Garovic VD, Alexander BT. Human Umbilical Venous Endothelial Cells: Early Predictors of Cardiovascular Risk in Offspring? Hypertension 2019; 74:32-34. [PMID: 31154902 DOI: 10.1161/hypertensionaha.119.12652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Jane F Reckelhoff
- From the Departments of Cell and Molecular Biology (J.F.R.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| | - Babbette LaMarca
- Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| | - Vesna D Garovic
- Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.).,Departments of Internal Medicine (V.G.), Mayo Clinic, Rochester, MN.,Hypertension and Nephrology (V.G.), Mayo Clinic, Rochester, MN
| | - Barbara T Alexander
- Physiology and Biophysics (B.T.A.), University of Mississippi Medical Center Jackson, MS.,Mississippi Center of Excellence in Perinatal Research (J.F.R., B.L., V.G., B.T.A.)
| |
Collapse
|
36
|
Zhou C, Yan Q, Zou QY, Zhong XQ, Tyler CT, Magness RR, Bird IM, Zheng J. Sexual Dimorphisms of Preeclampsia-Dysregulated Transcriptomic Profiles and Cell Function in Fetal Endothelial Cells. Hypertension 2019; 74:154-163. [PMID: 31154903 DOI: 10.1161/hypertensionaha.118.12569] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preeclampsia impairs fetoplacental vascular function and increases risks of adult-onset cardiovascular disorders in children born to preeclamptic mothers, implicating that preeclampsia programs fetal vasculature in utero. However, the underlying mechanisms remain elusive. We hypothesize that preeclampsia alters fetal endothelial gene expression and disturbs cytokines- and growth factors-induced endothelial responses. RNA sequencing analysis was performed on unpassaged human umbilical vein endothelial cells (HUVECs) from normotensive and preeclamptic pregnancies. Functional assays for endothelial monolayer integrity, proliferation, and migration were conducted on passage 1 HUVECs from normotensive and preeclamptic pregnancies. Compared with normotensive cells, 926 and 172 genes were dysregulated in unpassaged female and male HUVECs from preeclamptic pregnancies, respectively. Many of these preeclampsia-dysregulated genes are associated with cardiovascular diseases (eg, heart failure) and endothelial function (eg, cell migration, calcium signaling, and endothelial nitric oxide synthase signaling). TNF (tumor necrosis factor)-α-, TGF (transforming growth factor)-β1-, FGF (fibroblast growth factor)-2-, and VEGFA (vascular endothelial growth factor A)-regulated gene networks were differentially disrupted in unpassaged female and male HUVECs from preeclamptic pregnancies. Moreover, preeclampsia decreased endothelial monolayer integrity in responses to TNF-α in both female and male HUVECs. Preeclampsia decreased TGF-β1-strengthened monolayer integrity in female HUVECs, whereas it enhanced FGF-2-strengthened monolayer integrity in male HUVECs. Preeclampsia promoted TNF-α-, TGF-β1-, and VEGFA-induced cell proliferation in female, but not in male HUVECs. Preeclampsia inhibited TNF-α-induced cell migration in female HUVECs, but had an opposite effect on male HUVECs. In conclusion, preeclampsia differentially dysregulates cardiovascular diseases- and endothelial function-associated genes/pathways in female and male fetal endothelial cells in association with the sexual dimorphisms of preeclampsia-dysregulated fetal endothelial function.
Collapse
Affiliation(s)
- Chi Zhou
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.)
| | - Qin Yan
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.).,Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China (Q.Y.)
| | - Qing-Yun Zou
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.)
| | - Xin-Qi Zhong
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.).,Department of Pediatrics, the 3rd Affiliated Hospital of Guangzhou Medical University, Guangdong, China (X.-Q.Z.)
| | - Chanel T Tyler
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.)
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, University of South Florida, Tampa (R.R.M.)
| | - Ian M Bird
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.)
| | - Jing Zheng
- From the Department of Obstetrics and Gynecology, University of Wisconsin-Madison (C.Z., Q.Y., Q.-Y.Z., X.-Q.Z., C.T.T., I.M.B., J.Z.).,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (J.Z.)
| |
Collapse
|
37
|
Witt E, Lorenz M, Völker U, Stangl K, Hammer E, Stangl V. Sex-specific differences in the intracellular proteome of human endothelial cells from dizygotic twins. J Proteomics 2019; 201:48-56. [PMID: 30951907 DOI: 10.1016/j.jprot.2019.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/11/2019] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
Differences between men and women are being continuously identified in many human diseases. The underlying reasons are not yet fully understood. Beside the influence of endogenous hormones and life style, intrinsic sex-specific dimorphisms at the cellular level may also play a role. HUVECs from twin pairs of opposite sex provide an excellent tool to address the question of sex-specific differences at the molecular level. We compared for the first time protein levels of male and female HUVECs from dizygotic twins using a proteomic approach. To investigate differences under basal and stress conditions, cells were either left untreated or wounded and serum starved for different time points. Approximately 10% of all proteins monitored showed significant sexual dimorphisms in their level under the different conditions tested. The majority of the proteins displayed a higher abundance in female cells. The magnitude of the difference in protein levels between male and female cells was rather small. The most prominent differences throughout all conditions were observed for several X-chromosome encoded proteins with higher levels in female (UBA1, HDHD1) or in male cells (G6PD). Proteins involved in basic cellular processes, such as gene expression and translation (e.g. HMGN1, SRP54) displayed sex-specific levels in particular conditions only. SIGNIFICANCE: This study provides novel insights into sexual dimorphic protein levels in HUVECs from twin pairs of the opposite sex. The findings identify proteins with sex-specific differences in their levels under different cell culture conditions. The study also highlights the presence of X-chromosome encoded proteins escaping X-chromosomal inactivation. The results emphasize the need to consider the cellular sex of male and female HUVECs in in vitro experiments.
Collapse
Affiliation(s)
- Eric Witt
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany
| | - Mario Lorenz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany
| | - Karl Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany.
| | - Verena Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
38
|
Olmos-Ortiz A, García-Quiroz J, Halhali A, Avila E, Zaga-Clavellina V, Chavira-Ramírez R, García-Becerra R, Caldiño-Soto F, Larrea F, Díaz L. Negative correlation between testosterone and TNF-α in umbilical cord serum favors a weakened immune milieu in the human male fetoplacental unit. J Steroid Biochem Mol Biol 2019; 186:154-160. [PMID: 30359690 DOI: 10.1016/j.jsbmb.2018.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/10/2018] [Accepted: 10/18/2018] [Indexed: 12/27/2022]
Abstract
Clinical and epidemiological evidence supports that pregnancies carrying a male fetus are more vulnerable to infections and preterm birth, probably due to testosterone immunosuppressive properties. In human placentas, testosterone lowers the expression of CYP27B1, the vitamin D (VD)-activating enzyme, diminishing cathelicidin synthesis, a potent VD-dependent antimicrobial peptide (AMP). VD also stimulates other AMPs, including defensins. To get insights into the increased male vulnerability mechanisms, we investigated the relationship between fetal sex and the immunoendocrine milieu at the fetoplacental unit. For this, umbilical vein serum and placental samples were collected from healthy newborns. In males' serum, testosterone levels were significantly higher and negatively associated with TNF-α, a cytokine that strengthens the immune response. Males showed lower serum TNF-α and increased levels and gene expression of the immunosuppressive cytokine IL-10. Only in female samples there was a positive association (P < 0.05) between AMPs and both TNF-α and CYP27B1 and between 25-hydroxyvitamin D3 and IL-1β serum levels. Accordingly, VD-metabolites (25-hydroxyvitamin D3, calcitriol) significantly stimulated IL-1β gene expression in cultured trophoblasts. Interestingly, IL-1β mRNA correlated positively with defensins (P < 0.05) in males, but not with cathelicidin expression, which was significantly diminished in comparison to females. Our data suggest that high umbilical serum testosterone and IL-10 in males could explain reduced TNF-α levels and lack of association between VD-dependent innate immunity markers and proinflammatory cytokines expression in the fetoplacental unit. Altogether, our observations imply a restricted basal immune milieu in males compared to females, which may help understand the higher male susceptibility to adverse perinatal outcomes.
Collapse
Affiliation(s)
- Andrea Olmos-Ortiz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico; Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales No. 800, Lomas de Virreyes, C.P. 11000, Ciudad de México, Mexico
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico
| | - Ali Halhali
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico
| | - Euclides Avila
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico
| | - Verónica Zaga-Clavellina
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales No. 800, Lomas de Virreyes, C.P. 11000, Ciudad de México, Mexico
| | - Roberto Chavira-Ramírez
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico
| | - Rocío García-Becerra
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico; Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, Coyoacán 04360, Ciudad de México, Mexico
| | - Felipe Caldiño-Soto
- Jefatura de UTQ, UMAE Hospital de Gineco Obstetricia No. 4 "Luis Castelazo Ayala", IMSS, Av. Río Magdalena No. 289, Tizapán San Angel, C.P. 01090, Ciudad de México, Mexico
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, C.P. 14080, Ciudad de México, Mexico.
| |
Collapse
|
39
|
Micromanaging human placental function: differential microRNA expression in feto-placental endothelial cells of gestational diabetes pregnancies. Clin Sci (Lond) 2019; 133:315-319. [PMID: 30683714 DOI: 10.1042/cs20180901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022]
Abstract
Healthy development in utero relies on the appropriate exchange of nutrients and other signalling between the maternal and fetal circulations. Disruption to this fine balance is associated with several pregnancy and adverse birth outcomes, including gestational diabetes mellitus (GDM). This is a complex condition influenced by genetic, environment and potentially epigenetic factors in association with a range of altered developmental outcomes. A recent study, published in Clinical Science, explores miRNAs as a molecular mechanism underpinning the altered function of placental endothelial cells in GDM pregnancies.
Collapse
|
40
|
Gaba A, Mairhofer M, Zhegu Z, Leditznig N, Szabo L, Tschugguel W, Schneeberger C, Yotova I. Testosterone induced downregulation of migration and proliferation in human Umbilical Vein Endothelial Cells by Androgen Receptor dependent and independent mechanisms. Mol Cell Endocrinol 2018; 476:173-184. [PMID: 29777728 DOI: 10.1016/j.mce.2018.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022]
Abstract
Recent research has emphasized the potential unfavorable effects of declining testosterone (T) levels in men and the putative beneficial effect of androgen therapy in select women. Some controversy surrounding the mechanism of action and the effects of T on endothelium remains. In this study, we evaluated the mechanism of T action on pooled primary Human Umbilical Vein Endothelial Cells (HUVEC) of mixed gender by focusing on two important processes, proliferation and migration. In our in vitro model system, we found that only the supra-physiological dose of T affected these two processes irrespective of the ratio of male to female cells in the pools. At a concentration of 1 μM, T downregulated the proliferation of HUVEC by inducing arrest in the G1 cell cycle phase in an Androgen Receptor (AR)-independent manner. We show that treatment with 1 μM T also induced downregulation of HUVEC migration. This process was AR-dependent and was associated with persistent phosphorylation of ezrin, radixin and moesin. Regardless of the mechanism of action, the treatment of HUVEC with both supra- and physiological doses of T was associated with posttranscriptional stabilization of the AR upon ligand binding.
Collapse
Affiliation(s)
- Aulona Gaba
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | | | - Zyhdi Zhegu
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Nadja Leditznig
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Ladislaus Szabo
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Walter Tschugguel
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Christian Schneeberger
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Iveta Yotova
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Rudnicki M, Abdifarkosh G, Rezvan O, Nwadozi E, Roudier E, Haas TL. Female Mice Have Higher Angiogenesis in Perigonadal Adipose Tissue Than Males in Response to High-Fat Diet. Front Physiol 2018; 9:1452. [PMID: 30405427 PMCID: PMC6206240 DOI: 10.3389/fphys.2018.01452] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/25/2018] [Indexed: 01/21/2023] Open
Abstract
Background: Impaired capillary growth (angiogenesis) in skeletal muscle and adipose tissue contributes to the development of metabolic disorders in obese males. This association remains unexplored in females, despite mounting evidence that endothelial cells have sex-specific transcriptional profiles. Therefore, herein we assessed whether males and females show distinct angiogenic capacities in response to diet-induced obesity. Methods: Age-matched male and female mice were fed normal chow or high-fat obesogenic diets for 16 weeks. At the end of diet period, systemic glucose disposal was assessed as well as insulin sensitivity of skeletal muscle and visceral adipose tissue. Capillary content and the expression of angiogenic regulators were also evaluated in these tissues. Results: When placed on a high-fat diet, female mice gained less weight than males and showed a metabolic phenotype similar to NC-fed mice, contrasting with the impaired whole-body glucose metabolism observed in high-fat-fed males. However, high-fat-feeding elevated serum lipid levels similarly in male and female mice. Although skeletal muscle of high-fat-fed female mice had higher insulin sensitivity than male counterparts, no sex difference was detected in muscle capillarization. Metabolic functions of perigonadal white adipose tissue (pgWAT) were retained in high-fat-fed females, as evidenced by smaller adipocytes with preserved insulin sensitivity, greater responsiveness to isoproterenol, higher expression of Adiponectin and a lower ratio of Leptin:Adiponectin mRNA. An enhanced browning phenotype was detected in HF-fed female adipocytes with upregulation of Ucp1 expression. PgWAT from high-fat-fed females also showed augmented capillary number and expression of endothelial cell markers, which was associated with elevated mRNA levels of pro-angiogenic mediators, including vascular endothelial growth factor A (Vegfa) and its receptor (Vegfr2), the Notch ligand Jagged-1 (Jag1) and Angiopoietin-2 (Angpt2). Conclusion: Taken together, our findings provide novel evidence that visceral adipose tissue of female mice display greater levels of pro-angiogenic factors and vascularity than males in response to high-fat diet. This phenotype is associated with preserved metabolic homeostasis at both tissue and systemic levels. Our study discloses that a thus-far-unappreciated sex-specific difference in the regulation of adipose angiogenesis may contribute to an individual's susceptibility to developing adipose dysfunction and obesity-related metabolic disturbances.
Collapse
Affiliation(s)
- Martina Rudnicki
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Ghoncheh Abdifarkosh
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Omid Rezvan
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Emmanuel Nwadozi
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Emilie Roudier
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Tara L Haas
- Angiogenesis Research Group, School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
42
|
Xue XT, Zhang T, Cui SJ, He DQ, Wang XD, Yang RL, Liu DW, Liu Y, Gan YH, Kou XX, Zhou YH. Sexual dimorphism of estrogen-sensitized synoviocytes contributes to gender difference in temporomandibular joint osteoarthritis. Oral Dis 2018; 24:1503-1513. [PMID: 29806726 DOI: 10.1111/odi.12905] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/25/2018] [Accepted: 05/15/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Temporomandibular joint osteoarthritis (TMJOA) is approximately twice as prevalent in women than in men. Synoviocytes are believed to play a critical role in joint inflammation. However, it is unknown whether synoviocytes from different genders possess sexual dimorphisms that contribute to female-predominant TMJOA. MATERIALS AND METHODS Freund's complete adjuvant combined with monosodium iodoacetate was used to induce TMJOA in female and male rats. Histologic and radiographic features were used to evaluate TMJOA. The expression of CD68, MCP-1, iNOS, and IL-1β was detected by immunohistochemistry and real-time PCR. Primary fibroblast-like synoviocytes (FLSs) isolated from the synovial membrane of female and male rats were used for in vitro experiments. RESULTS Female rats showed aggravated TMJOA features as compared to male rats. Increased expression of iNOS and IL-1β was detected in synovial membrane from female TMJOA rats as compared to male rats. Furthermore, greater amounts of CD68-positive macrophage infiltration and increased MCP-1 expression around the synovial membrane were detected in female TMJOA rats compared to males. Primary cultured FLSs from female rats showed higher sensitivity to TNF-α treatment and recruited increased macrophage migration than male FLSs. More important, ovariectomy (OVX) by ablation in female rats repressed the sensitivity of female FLSs to TNF-α treatment due to the loss of estrogen production. Blockage of the estrogen receptor repressed estrogen-potentiated TNF-α-induced pro-inflammatory cytokine expression in OVX-FLSs. Moreover, the injection of estrogen receptor antagonists relieved the cartilage destruction and bone deterioration of TMJOA in female rats. CONCLUSION Estrogen-sensitized synoviocytes in female rats may contribute to gender differences in the incidence and progression of TMJOA.
Collapse
Affiliation(s)
- Xin-Tong Xue
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ting Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Sheng-Jie Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Dan-Qing He
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xue-Dong Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Rui-Li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Da-Wei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ye-Hua Gan
- Center for Temporomandibular Disorders and Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao-Xing Kou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Center for Craniofacial Stem Cell Research and Regeneration, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
43
|
Zhang Y, Dong X, Shirazi J, Gleghorn JP, Lingappan K. Pulmonary endothelial cells exhibit sexual dimorphism in their response to hyperoxia. Am J Physiol Heart Circ Physiol 2018; 315:H1287-H1292. [PMID: 30095998 PMCID: PMC6415740 DOI: 10.1152/ajpheart.00416.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abnormal pulmonary vascular development is a critical factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Despite the well-established sex-specific differences in the incidence of BPD, the molecular mechanism(s) behind these are not completely understood. Exposure to a high concentration of oxygen (hyperoxia) contributes to BPD and creates a profibrotic environment in the lung. Our objective was to elucidate the sex-specific differences in neonatal human pulmonary microvascular endothelial cells (HPMECs) in normoxic and hyperoxic conditions, including the propensity for endothelial-to-mesenchymal transition. HPMECs (18- to 24-wk gestation donors, 6 male donors and 5 female donors) were subjected to hyperoxia (95% O2 and 5% CO2) or normoxia (air and 5% CO2) up to 72 h. We assessed cell migration and angiogenesis at baseline. Cell proliferation, viability, and expression of endothelial (CD31) and fibroblast markers (α-smooth muscle actin) were measured upon exposure to hyperoxia. Female HPMECs had significantly higher cell migration when assessed by the wound healing assay (40.99 ± 4.4%) compared with male HPMECs (14.76 ± 3.7%) and showed greater sprouting (1710 ± 962 μm in female cells vs. 789 ± 324 in male cells) compared with male endothelial cells in normoxia. Hyperoxia exposure decreased cell viability (by 9.8% at 48 h and 11.7% at 72 h) and proliferation (by 26.7% at 72 h) markedly in male HPMECs, whereas viability was sustained in female endothelial cells. There was greater expression of α-smooth muscle actin (2.5-fold) and decreased expression (5-fold) of CD31 in male HPMECs upon exposure to hyperoxia. The results indicate that cellular sex affects response in HPMECs in normoxia and hyperoxia. NEW & NOTEWORTHY Cellular sex affects response in human neonatal pulmonary microvascular endothelial cells in normoxia and hyperoxia. Under normoxic conditions, female human neonatal pulmonary microvascular endothelial cells display greater migration and angiogenic sprouting compared with male endothelial cells. Compared with female endothelial cells, hyperoxia exposure decreased cell viability and proliferation and increased α-smooth muscle actin and decreased CD31 expression in male endothelial cells, indicating an increased endothelial-mesenchymal transition.
Collapse
Affiliation(s)
- Yuhao Zhang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Xiaoyu Dong
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware , Newark, Delaware
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware , Newark, Delaware
| | - Krithika Lingappan
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
44
|
Huxley VH, Kemp SS, Schramm C, Sieveking S, Bingaman S, Yu Y, Zaniletti I, Stockard K, Wang J. Sex differences influencing micro- and macrovascular endothelial phenotype in vitro. J Physiol 2018; 596:3929-3949. [PMID: 29885204 DOI: 10.1113/jp276048] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Endothelial dysfunction is an early hallmark of multiple disease states that also display sex differences with respect to age of onset, frequency and severity. Results of in vivo studies of basal and stimulated microvascular barrier function revealed sex differences that are difficult to ascribe to specific cells or environmental factors. The present study evaluated endothelial cells (EC) isolated from macro- and/or microvessels of reproductively mature rats under the controlled conditions of low-passage culture aiming to test the assumption that EC phenotype would be sex independent. The primary finding was that EC, regardless of where they are derived, retain a sex-bias in low-passage culture, independent of varying levels of reproductive hormones. The implications of the present study include the fallacy of expecting a universal set of mechanisms derived from study of EC from one sex and/or one vascular origin to apply uniformly to all EC under unstimulated conditions, and no less in disease. ABSTRACT Vascular endothelial cells (EC) are heterogeneous with respect to phenotype, reflecting at least the organ of origin, location within the vascular network and physical forces. As an independent influence on EC functions in health or aetiology, susceptibility, and progression of dysfunction in numerous disease states, sex has been largely ignored. The present study focussed on EC isolated from aorta (macrovascular) and skeletal muscle vessels (microvascular) of age-matched male and female rats under identical conditions of short-term (passage 4) culture. We tested the hypothesis that genomic sex would not influence endothelial growth, wound healing, morphology, lactate production, or messenger RNA and protein expression of key proteins (sex hormone receptors for androgen and oestrogens α and β; platelet endothelial cell adhesion molecule-1 and vascular endothelial cadherin mediating barrier function; αv β3 and N-cadherin influencing matrix interactions; intracellular adhesion molecule-1 and vascular cell adhesion molecule-1 mediating EC/white cell adhesion). The hypothesis was rejected because the EC origin (macro- vs. microvessel) and sex influenced multiple phenotypic characteristics. Statistical model analysis of EC growth demonstrated an hierarchy of variable importance, recapitulated for other phenotypic characteristics, with predictions assuming EC homogeneity < sex < vessel origin < sex and vessel origin. Furthermore, patterns of EC mRNA expression by vessel origin and by sex did not predict protein expression. Overall, the present study demonstrated that accurate assessment of sex-linked EC dysfunction first requires an understanding of EC function by position in the vascular tree and by sex. The results from a single EC tissue source/species/sex cannot provide universal insight into the mechanisms regulating in vivo endothelial function in health, and no less in disease.
Collapse
Affiliation(s)
- Virginia H Huxley
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Scott S Kemp
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Christine Schramm
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Steve Sieveking
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Susan Bingaman
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Yang Yu
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Isabella Zaniletti
- Department of Statistics, University of Missouri-Columbia, Columbia, MO, USA
| | - Kevin Stockard
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Jianjie Wang
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Biomedical Sciences, Missouri State University, Springfield, MO, USA
| |
Collapse
|
45
|
Vanetti C, Bifari F, Vicentini LM, Cattaneo MG. Fatty acids rather than hormones restore in vitro angiogenesis in human male and female endothelial cells cultured in charcoal-stripped serum. PLoS One 2017; 12:e0189528. [PMID: 29232396 PMCID: PMC5726635 DOI: 10.1371/journal.pone.0189528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/27/2017] [Indexed: 01/18/2023] Open
Abstract
Charcoal-stripped serum (CSS) is a well-accepted method to model effects of sex hormones in cell cultures. We have recently shown that human endothelial cells (ECs) fail to growth and to undergo in vitro angiogenesis when cultured in CSS. However, the mechanism(s) underlying the CSS-induced impairment of in vitro EC properties are still unknown. In addition, whether there is any sexual dimorphism in the CSS-induced EC phenotype remains to be determined. Here, by independently studying human male and female ECs, we found that CSS inhibited both male and female EC growth and in vitro angiogenesis, with a more pronounced effect on male EC sprouting. Reconstitution of CSS with 17-β estradiol, dihydrotestosterone, or the lipophilic thyroid hormone did not restore EC functions in both sexes. On the contrary, supplementation with palmitic acid or the acetyl-CoA precursor acetate significantly rescued the CSS-induced inhibition of growth and sprouting in both male and female ECs. We can conclude that the loss of metabolic precursors (e.g., fatty acids) rather than of hormones is involved in the impairment of in vitro proliferative and angiogenic properties of male and female ECs cultured with CSS.
Collapse
Affiliation(s)
- Claudia Vanetti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Lucia M. Vicentini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
- * E-mail:
| |
Collapse
|
46
|
Cattaneo MG, Vanetti C, Decimo I, Di Chio M, Martano G, Garrone G, Bifari F, Vicentini LM. Sex-specific eNOS activity and function in human endothelial cells. Sci Rep 2017; 7:9612. [PMID: 28852041 PMCID: PMC5575132 DOI: 10.1038/s41598-017-10139-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Clinical and epidemiological data show that biological sex is one of the major determinants for the development and progression of cardiovascular disease (CVD). Impaired endothelial function, characterized by an imbalance in endothelial Nitric Oxide Synthase (eNOS) activity, precedes and accelerates the development of CVD. However, whether there is any sexual dimorphism in eNOS activity and function in endothelial cells (ECs) is still unknown. Here, by independently studying human male and female ECs, we found that female ECs expressed higher eNOS mRNA and protein levels both in vitro and ex vivo. The increased eNOS expression was associated to higher enzymatic activity and nitric oxide production. Pharmacological and genetic inhibition of eNOS affected migratory properties only in female ECs. In vitro angiogenesis experiments confirmed that sprouting mostly relied on eNOS-dependent migration in female ECs. At variance, capillary outgrowth from male ECs was independent of eNOS activity but required cell proliferation. In this study, we found sex-specific differences in the EC expression, activity, and function of eNOS. This intrinsic sexual dimorphism of ECs should be further evaluated to achieve more effective and precise strategies for the prevention and therapy of diseases associated to an impaired endothelial function such as CVD and pathological angiogenesis.
Collapse
Affiliation(s)
- Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy.
| | - Claudia Vanetti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | - Ilaria Decimo
- Department of Diagnostics and Public Health, Università di Verona, 37134, Verona, Italy
| | - Marzia Di Chio
- Department of Diagnostics and Public Health, Università di Verona, 37134, Verona, Italy
| | | | - Giulia Garrone
- Fondazione IRCCS, Istituto Nazionale dei Tumori, 20133, Milano, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | - Lucia Maria Vicentini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy.
| |
Collapse
|
47
|
Keeley TP, Siow RCM, Jacob R, Mann GE. A PP2A-mediated feedback mechanism controls Ca 2+-dependent NO synthesis under physiological oxygen. FASEB J 2017; 31:5172-5183. [PMID: 28760745 PMCID: PMC5690389 DOI: 10.1096/fj.201700211r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
Intracellular O2 is a key regulator of NO signaling, yet most in vitro studies are conducted in atmospheric O2 levels, hyperoxic with respect to the physiologic milieu. We investigated NO signaling in endothelial cells cultured in physiologic (5%) O2 and stimulated with histamine or shear stress. Culture of cells in 5% O2 (>5 d) decreased histamine- but not shear stress–stimulated endothelial (e)NOS activity. Unlike cells adapted to a hypoxic environment (1% O2), those cultured in 5% O2 still mobilized sufficient Ca2+ to activate AMPK. Enhanced expression and membrane targeting of PP2A-C was observed in 5% O2, resulting in greater interaction with eNOS in response to histamine. Moreover, increased dephosphorylation of eNOS in 5% O2 was Ca2+-sensitive and reversed by okadaic acid or PP2A-C siRNA. The present findings establish that Ca2+ mobilization stimulates both NO synthesis and PP2A-mediated eNOS dephosphorylation, thus constituting a novel negative feedback mechanism regulating eNOS activity not present in response to shear stress. This, coupled with enhanced NO bioavailability, underpins differences in NO signaling induced by inflammatory and physiologic stimuli that are apparent only in physiologic O2 levels. Furthermore, an explicit delineation between physiologic normoxia and genuine hypoxia is defined here, with implications for our understanding of pathophysiological hypoxia.—Keeley, T. P., Siow, R. C. M., Jacob, R., Mann, G. E. A PP2A-mediated feedback mechanism controls Ca2+-dependent NO synthesis under physiological oxygen.
Collapse
Affiliation(s)
- Thomas P Keeley
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Richard C M Siow
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ron Jacob
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Giovanni E Mann
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
48
|
Benn A, Hiepen C, Osterland M, Schütte C, Zwijsen A, Knaus P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence. FASEB J 2017; 31:4720-4733. [PMID: 28733457 PMCID: PMC5636702 DOI: 10.1096/fj.201700193rr] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 01/04/2023]
Abstract
Before the onset of sprouting angiogenesis, the endothelium is prepatterned for the positioning of tip and stalk cells. Both cell identities are not static, as endothelial cells (ECs) constantly compete for the tip cell position in a dynamic fashion. Here, we show that both bone morphogenetic protein 2 (BMP2) and BMP6 are proangiogenic in vitro and ex vivo and that the BMP type I receptors, activin receptor-like kinase 3 (ALK3) and ALK2, play crucial and distinct roles in this process. BMP2 activates the expression of tip cell-associated genes, such as delta-like ligand 4 (DLL4) and kinase insert domain receptor (KDR), and p38-heat shock protein 27 (HSP27)-dependent cell migration, thereby generating tip cell competence. Whereas BMP6 also triggers collective cell migration via the p38-HSP27 signaling axis, BMP6 induces in addition SMAD1/5 signaling, thereby promoting the expression of stalk cell-associated genes, such as hairy and enhancer of split 1 (HES1) and fms-like tyrosine kinase 1 (FLT1). Specifically, ALK3 is required for sprouting from HUVEC spheroids, whereas ALK2 represses sprout formation. We demonstrate that expression levels and respective complex formation of BMP type I receptors in ECs determine stalk vs. tip cell identity, thus contributing to endothelial plasticity during sprouting angiogenesis. As antiangiogenic monotherapies that target the VEGF or ALK1 pathways have not fulfilled efficacy objectives in clinical trials, the selective targeting of the ALK2/3 pathways may be an attractive new approach.-Benn, A., Hiepen, C., Osterland, M., Schütte, C., Zwijsen, A., Knaus, P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.,Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Marc Osterland
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - Christof Schütte
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - An Zwijsen
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; .,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
49
|
Liu P, Li X, Song F, Li P, Wei J, Yan Q, Xu X, Yang J, Li C, Fu X. Testosterone promotes tube formation of endothelial cells isolated from veins via activation of Smad1 protein. Mol Cell Endocrinol 2017; 446:21-31. [PMID: 28167128 DOI: 10.1016/j.mce.2017.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/11/2017] [Accepted: 02/02/2017] [Indexed: 01/03/2023]
Abstract
Testosterone (T) deficiency is positively correlated with the increased incidence of cardiovascular disease. However, the effects of T on vascular endothelial cells remain obscure. Tube formation capacity is critical for vascular regeneration/repair and Smad1 plays an important role in these events. In this study, we investigated the effects of T on Smad1 activation and tube formation of cultured human umbilical endothelial cells (HUVECs). Our results showed that T rapidly increased endothelial Smad1 phosphorylation. This effect was mimicked by cell-impermeable T-BSA conjugates and was not altered by transcriptional inhibitor actinomycin D or translational inhibitor cycloheximide. T-induced Smad1 phosphorylation was blocked by ERK1/2 and c-Src inhibitors or their specific siRNAs, while it was reinforced by ERK1/2 or c-Src overexpression. Indeed, T rapidly activated ERK1/2 and c-Src signalings and c-Src was confirmed as the upstream of ERK1/2. Moreover, caveolae disruptor methyl-β-cyclodextrin (β-MCD) blocked Smad1 activation induced by T. The association of caveolin-1 with androgen receptor (AR) or c-Src was detected by immunoprecipitation and it was significantly increased by rapid T stimulation. Furthermore, fractional analysis showed that AR and c-Src were expressed in caveolae-enriched membrane fractions. T promoted tube formation of HUVECs, which was blocked by c-Src and ERK1/2 inhibitors or by the knockdown of Smad1. In conclusion, T increased tube formation of endothelial cells isolated from veins by stimulating Smad1 phosphorylation in a nongenomic manner, which was mediated by signals from AR/c-Src located in caveolae to ERK1/2 cascade. These findings may shed new light on the relevance of T to its vascular functions.
Collapse
Affiliation(s)
- Pei Liu
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Xiaosa Li
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Fuhu Song
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Ping Li
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jinzhi Wei
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Qing Yan
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xingyan Xu
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jun Yang
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chuanxiang Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China.
| | - Xiaodong Fu
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
50
|
Differential sex-specific effects of oxygen toxicity in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2017; 486:431-437. [PMID: 28315681 DOI: 10.1016/j.bbrc.2017.03.058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/14/2017] [Indexed: 11/21/2022]
Abstract
Despite the well-established sex-specific differences in the incidence of bronchopulmonary dysplasia (BPD), the molecular mechanism(s) behind these are not completely understood. Pulmonary angiogenesis is critical for alveolarization and arrest in vascular development adversely affects lung development. Human neonatal umbilical vein endothelial cells (HUVECs) provide a robust in vitro model for the study of endothelial cell physiology and function. Male and Female HUVECs were exposed to room air (21% O2, 5% CO2) or hyperoxia (95% O2, 5% CO2) for up to 72 h. Cell viability, proliferation, H2O2 production and angiogenesis were analyzed. Sex-specific differences in the expression of VEGFR2 and modulation of NF-kappa B pathway were measured. Male HUVECs have decreased survival, greater oxidative stress and impairment in angiogenesis compared to similarly exposed female cells. There is differential expression of VEGFR2 between male and female HUVECs and greater activation of the NF-kappa B pathway in female HUVECs under hyperoxic conditions. The results indicate that sex differences exist between male and female HUVECs in vitro after hyperoxia exposure. Since endothelial dysfunction has a major role in the pathogenesis of BPD, these differences could explain in part the mechanisms behind sex-specific differences in the incidence of this disease.
Collapse
|