1
|
Cassavaugh J, Longhi MS, Robson SC. Impact of Estrogen on Purinergic Signaling in Microvascular Disease. Int J Mol Sci 2025; 26:2105. [PMID: 40076726 PMCID: PMC11900469 DOI: 10.3390/ijms26052105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Microvascular ischemia, especially in the heart and kidneys, is associated with inflammation and metabolic perturbation, resulting in cellular dysfunction and end-organ failure. Heightened production of adenosine from extracellular nucleotides released in response to inflammation results in protective effects, inclusive of adaptations to hypoxia, endothelial cell nitric oxide release with the regulation of vascular tone, and inhibition of platelet aggregation. Purinergic signaling is modulated by ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1)/CD39, which is the dominant factor dictating vascular metabolism of extracellular ATP to adenosine throughout the cardiovascular tissues. Excess levels of extracellular purine metabolites, however, have been associated with metabolic and cardiovascular diseases. Physiological estrogen signaling is anti-inflammatory with vascular protective effects, but pharmacological replacement use in transgender and postmenopausal individuals is associated with thrombosis and other side effects. Crucially, the loss of this important sex hormone following menopause or with gender reassignment is associated with worsened pro-inflammatory states linked to increased oxidative stress, myocardial fibrosis, and, ultimately, diastolic dysfunction, also known as Yentl syndrome. While there is a growing body of knowledge on distinctive purinergic or estrogen signaling and endothelial health, much less is known about the relationships between the two signaling pathways. Continued studies of the interactions between these pathways will allow further insight into future therapeutic targets to improve the cardiovascular health of aging women without imparting deleterious side effects.
Collapse
Affiliation(s)
- Jessica Cassavaugh
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (M.S.L.); (S.C.R.)
| | | | | |
Collapse
|
2
|
Jain K, Tyagi T, Gu SX, Faustino EVS, Hwa J. Demographic diversity in platelet function and response to antiplatelet therapy. Trends Pharmacol Sci 2025; 46:78-93. [PMID: 39672782 PMCID: PMC11710996 DOI: 10.1016/j.tips.2024.11.005] [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: 09/23/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 12/15/2024]
Abstract
Recent studies have highlighted the complexity of platelet biology, revealing their diverse roles beyond hemostasis. Pathological platelet activation is now recognized as a key contributor to thrombosis and inflammation that are both central to cardiovascular disease (CVD). Emerging research emphasizes the significant impact of demographic factors - such as age, sex, race, and ethnicity - on CVD risk and responses to antiplatelet therapies. These population-based differences, shaped by genetic and non-genetic factors, highlight the need for reevaluation of antiplatelet strategies. We address current knowledge and emphasize the pressing need for further research into platelet biology and cardiovascular outcomes across diverse populations. In this review we advocate for tailored therapeutic approaches in CVD based on the recent demographic-focused findings.
Collapse
Affiliation(s)
- Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Yale Cooperative Center of Excellence in Hematology, Yale School of Medicine, New Haven, CT, USA.
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Yale Cooperative Center of Excellence in Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cooperative Center of Excellence in Hematology, Yale School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - E Vincent S Faustino
- Yale Cooperative Center of Excellence in Hematology, Yale School of Medicine, New Haven, CT, USA; Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Yale Cooperative Center of Excellence in Hematology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Battipaglia C, Genazzani AD, Nappi RE, La Marca A. Insights on estetrol, the native estrogen: from contraception to hormone replacement therapy. Minerva Obstet Gynecol 2024; 76:590-603. [PMID: 39283289 DOI: 10.23736/s2724-606x.24.05594-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Estetrol (E4) is a natural estrogen that has recently emerged as new option for contraception and hormone replacement therapy (HRT). Unlike other estrogens, E4 primarily stimulates nuclear estrogen receptor alpha (ERα) and does not activate membrane ERα. For this reason, this novel estrogen has tissue-specific effects across various organs such as liver, vascular endothelium, mammary glands, brain, vagina, and uterus. The selective activation of the nuclear ERα results in distinct pharmacological properties that contribute to its unique therapeutic profile. Moreover, E4 shows minimal interaction with the hepatic cytochrome P450 enzyme system, leading to a favorable pharmacokinetic profile and a reduced potential for drug-drug interactions. Currently, E4 is commercially available in combination with drospirenone as a combined oral contraceptive and its application in HRT is undergoing late-stage clinical development. Many studies have demonstrated that E4 has a lower impact on hemostatic and metabolic parameters compared to other estrogens, potentially reducing the risk of adverse effects commonly associated with hormonal therapies such as thromboembolic events or dyslipidemia. Beyond its role in contraception and HRT, E4 shows promising therapeutic potential in other medical fields, including neuroprotection in neonatal hypoxic-ischemic encephalopathy, enhancement of hematopoietic stem cell transplantation outcomes and prostate cancer management. This review synthesizes the latest evidence on E4 primarily focusing on its pharmacological characteristics and clinical applications. The findings suggest that E4 versatility and peculiar mechanism of action may represent an important therapeutic option for a broad spectrum of medical conditions.
Collapse
Affiliation(s)
- Christian Battipaglia
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy -
| | - Alessandro D Genazzani
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS S. Matteo Foundation, Pavia, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Kielb J, Saffak S, Weber J, Baensch L, Shahjerdi K, Celik A, Farahat N, Riek S, Chavez-Talavera O, Grandoch M, Polzin A, Kelm M, Dannenberg L. Transformation or replacement - Effects of hormone therapy on cardiovascular risk. Pharmacol Ther 2024; 254:108592. [PMID: 38286163 DOI: 10.1016/j.pharmthera.2024.108592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2024]
Abstract
Hormone therapy (HT) is important and frequently used both regarding replacement therapy (HRT) and gender affirming therapy (GAHT). While HRT has been effective in addressing symptoms related to hormone shortage, several side effects have been described. In this context, there are some studies that show increased cardiovascular risk. However, there are also studies reporting protective aspects of HT. Nevertheless, the exact impact of HT on cardiovascular risk and the underlying mechanisms remain poorly understood. This article explores the relationship between diverse types of HT and cardiovascular risk, focusing on mechanistic insights of the underlying hormones on platelet and leukocyte function as well as on effects on endothelial and adipose tissue cells.
Collapse
Affiliation(s)
- Julia Kielb
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Süreyya Saffak
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Jessica Weber
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Leonard Baensch
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Khatereh Shahjerdi
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Aylin Celik
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Nora Farahat
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Sally Riek
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Oscar Chavez-Talavera
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Maria Grandoch
- Institute for Translational Pharmacology, Medical Faculty and University Hospital of Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany.
| |
Collapse
|
5
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [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: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
6
|
Ghatge M, Nayak MK, Flora GD, Kumskova M, Jain A, Patel RB, Lin Z, Usachev YM, Chauhan AK. Mitochondrial calcium uniporter b deletion inhibits platelet function and reduces susceptibility to arterial thrombosis. J Thromb Haemost 2023; 21:2163-2174. [PMID: 37061131 DOI: 10.1016/j.jtha.2023.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Mitochondrial calcium uniporter b (MCUb) is a negative regulator of the mitochondrial calcium uniporter (MCU) and is known to limit mitochondrial calcium ion (Ca2+) uptake. The role of MCUb in platelet function remains unclear. OBJECTIVES Utilizing MCUb-/- mice, we examined the role of MCUb in regulating platelet function and thrombosis. METHODS Platelet activation was evaluated in agonist-induced standardized in vitro assays. Susceptibility to arterial thrombosis was evaluated in FeCl3 injury-induced carotid artery and laser injury-induced mesenteric artery thrombosis models. The glycolytic proton efflux rate and oxygen consumption rate were measured to evaluate aerobic glycolysis. RESULTS Upon stimulation, MCUb-/- platelets exhibited reduced cytoplasmic Ca2+ responses concomitant with increased mitochondrial Ca2+ uptake. MCUb-/- platelets displayed reduced agonist-induced platelet aggregation and spreading on fibrinogen and decreased α and dense-granule secretion and clot retraction. MCUb-/- mice were less susceptible to arterial thrombosis in FeCl3 injury-induced carotid and laser injury-induced mesenteric thrombosis models with unaltered tail bleeding time. In adoptive transfer experiments, thrombocytopenic hIL-4Rα/GPIbα-transgenic mice transfused with MCUb-/- platelets were less susceptible to FeCl3 injury-induced carotid thrombosis compared with hIL-4Rα/GPIbα-Tg mice transfused with wild type platelets, suggesting a platelet-specific role of MCUb in thrombosis. MCUb-/- stimulated platelets exhibited reduced glucose uptake, decreased glycolytic rate, and lowered pyruvate dehydrogenase phosphorylation, suggesting that mitochondrial Ca2+ mediates bioenergetic changes in platelets. CONCLUSION Our findings suggest that mitochondrial Ca2+ signaling and glucose oxidation are functionally linked in activated platelets and reveal a novel role of MCUb in platelet activation and arterial thrombosis.
Collapse
Affiliation(s)
- Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA.
| | - Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Mariia Kumskova
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Aditi Jain
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Rakesh B Patel
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Zhihong Lin
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Yuriy M Usachev
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
7
|
Bouck EG, Arvanitis M, Osburn WO, Sang Y, Reventun P, Ahmadzia HK, Smith NL, Lowenstein CJ, Wolberg AS. High risk oral contraceptive hormones do not directly enhance endothelial cell procoagulant activity in vitro. PLoS One 2023; 18:e0284333. [PMID: 37075041 PMCID: PMC10115293 DOI: 10.1371/journal.pone.0284333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/28/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Oral contraceptive (OC) use increases venous thromboembolism risk 2-5-fold. Procoagulant changes can be detected in plasma from OC users even without thrombosis, but cellular mechanisms that provoke thrombosis have not been identified. Endothelial cell (EC) dysfunction is thought to initiate venous thromboembolism. It is unknown whether OC hormones provoke aberrant procoagulant activity in ECs. OBJECTIVE Characterize the effect of high-risk OC hormones (ethinyl estradiol [EE] and drospirenone) on EC procoagulant activity and the potential interplay with nuclear estrogen receptors ERα and ERβ and inflammatory processes. METHODS Human umbilical vein and dermal microvascular ECs (HUVEC and HDMVEC, respectively) were treated with EE and/or drospirenone. Genes encoding the estrogen receptors ERα and ERβ (ESR1 and ESR2, respectively) were overexpressed in HUVEC and HDMVEC via lentiviral vectors. EC gene expression was assessed by RT-qPCR. The ability of ECs to support thrombin generation and fibrin formation was measured by calibrated automated thrombography and spectrophotometry, respectively. RESULTS Neither EE nor drospirenone, alone or together, changed expression of genes encoding anti- or procoagulant proteins (TFPI, THBD, F3), integrins (ITGAV, ITGB3), or fibrinolytic mediators (SERPINE1, PLAT). EE and/or drospirenone did not increase EC-supported thrombin generation or fibrin formation, either. Our analyses indicated a subset of individuals express ESR1 and ESR2 transcripts in human aortic ECs. However, overexpression of ESR1 and/or ESR2 in HUVEC and HDMVEC did not facilitate the ability of OC-treated ECs to support procoagulant activity, even in the presence of a pro-inflammatory stimulus. CONCLUSIONS The OC hormones EE and drospirenone do not directly enhance thrombin generation potential of primary ECs in vitro.
Collapse
Affiliation(s)
- Emma G. Bouck
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Marios Arvanitis
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - William O. Osburn
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yaqiu Sang
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Paula Reventun
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Homa K. Ahmadzia
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, George Washington University, Washington, DC, United States of America
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, United States of America
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, United States of America
| | - Charles J. Lowenstein
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
8
|
Chang S, Larsen OH, Hvas AM, Skakkebaek A, Gravholt CH, Münster AMB. Platelet aggregation in Klinefelter syndrome is not aggravated by testosterone replacement therapy: A longitudinal follow-up study. Andrology 2023; 11:456-463. [PMID: 36300871 DOI: 10.1111/andr.13330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Men with Klinefelter syndrome (KS) are routinely offered testosterone replacement therapy (TRT) suggested to potentially promote platelet aggregation and increase cardiovascular risk. OBJECTIVE We investigated platelet aggregation in men with KS before and during TRT. MATERIALS AND METHODS Forty-one adult men with KS participated, of which 20 had no history of TRT at baseline, with 15 completing follow-up after 18 months TRT. Further, we included 21 adult men with KS on long-term TRT (>10 years) and a male reference population. We assessed platelet impedance aggregometry using adenosine diphosphate (6.5 μM), thrombin-receptor-activating-peptide-6 (TRAP 32 μM), and arachidonic acid (ASPI 0.5 mM) as agonists in KS compared to a male reference population and stratified by route of TRT administration. RESULTS Platelet aggregation among men with KS at baseline or during TRT was not increased compared with the male reference population. For all three agonist, no change was seen in platelet aggregation in KS at follow-up compared with baseline (p ≥ 0.2). Platelet aggregation was not associated with total testosterone and furthermore, platelet count was not affected by treatment with testosterone. Men with KS treated with testosterone gel showed slightly increased TRAP- and ASPI-induced platelet aggregation compared with those treated with testosterone injection (p = 0.02 and p = 0.04, respectively). DISCUSSION AND CONCLUSIONS We observed normal platelet aggregation in men with KS before TRT and following both short and long term treatment. Our findings do not support an independent role of platelets in driving the cardiovascular risk in KS.
Collapse
Affiliation(s)
- Simon Chang
- Unit for Thrombosis Research, University of Southern Denmark, Esbjerg, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Ole Halfdan Larsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne Skakkebaek
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Højbjerg Gravholt
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
9
|
Li Z, Muench G, Goebel S, Uhland K, Wenhart C, Reimann A. Flow chamber staining modality for real-time inspection of dynamic phenotypes in multiple histological stains. PLoS One 2023; 18:e0284444. [PMID: 37141296 PMCID: PMC10159194 DOI: 10.1371/journal.pone.0284444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Traditional histological stains, such as hematoxylin-eosin (HE), special stains, and immunofluorescence (IF), have defined myriads of cellular phenotypes and tissue structures in a separate stained section. However, the precise connection of information conveyed by the various stains in the same section, which may be important for diagnosis, is absent. Here, we present a new staining modality-Flow chamber stain, which complies with the current staining workflow but possesses newly additional features non-seen in conventional stains, allowing for (1) quickly switching staining modes between destain and restain for multiplex staining in one single section from routinely histological preparation, (2) real-time inspecting and digitally capturing each specific stained phenotype, and (3) efficiently synthesizing graphs containing the tissue multiple-stained components at site-specific regions. Comparisons of its stains with those by the conventional staining fashions using the microscopic images of mouse tissues (lung, heart, liver, kidney, esophagus, and brain), involving stains of HE, Periodic acid-Schiff, Sirius red, and IF for Human IgG, and mouse CD45, hemoglobin, and CD31, showed no major discordance. Repetitive experiments testing on targeted areas of stained sections confirmed the method is reliable with accuracy and high reproducibility. Using the technique, the targets of IF were easily localized and seen structurally in HE- or special-stained sections, and the unknown or suspected components or structures in HE-stained sections were further determined in histological special stains or IF. By the technique, staining processing was videoed and made a backup for off-site pathologists, which facilitates tele-consultation or -education in current digital pathology. Mistakes, which might occur during the staining process, can be immediately found and amended accordingly. With the technique, a single section can provide much more information than the traditional stained counterpart. The staining mode bears great potential to become a common supplementary tool for traditional histopathology.
Collapse
|
10
|
Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. Int J Mol Sci 2022; 23:ijms232113176. [PMID: 36361963 PMCID: PMC9656618 DOI: 10.3390/ijms232113176] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.
Collapse
|
11
|
Wang ZP, Hua M, Jiu T, Ge RL, Bai Z. Biofunctional roles of estrogen in coronavirus disease 2019: Beyond a steroid hormone. Front Pharmacol 2022; 13:1003469. [PMID: 36339571 PMCID: PMC9626865 DOI: 10.3389/fphar.2022.1003469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/06/2022] [Indexed: 09/26/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), epidemic poses a major global public health threat with more than one million daily new infections and hundreds of deaths. To combat this global pandemic, efficient prevention and management strategies are urgently needed. Together with the main characteristics of COVID-19, impaired coagulation with dysfunctions of the immune response in COVID-19 pathophysiology causes high mortality and morbidity. From recent clinical observations, increased expression of specific types of estrogen appears to protect patients from SARS-CoV-2 infection, thereby, reducing mortality. COVID-19 severity is less common in women than in men, particularly in menopausal women. Furthermore, estrogen levels are negatively correlated with COVID-19 severity and mortality. These findings suggest that estrogen plays a protective role in the pathophysiology of COVID-19. In this review, we discuss the potential roles of estrogen in blocking the SARS-CoV-2 from invading alveolar cells and replicating, and summarize the potential mechanisms of anti-inflammation, immune modulation, reactive oxygen species resistance, anti-thrombosis, vascular dilation, and vascular endothelium protection. Finally, the potential therapeutic effects of estrogen against COVID-19 are reviewed. This review provides insights into the role of estrogen and its use as a potential strategy to reduce the mortality associated with COVID-19, and possibly other viral infections and discusses the possible challenges and pertinent questions.
Collapse
Affiliation(s)
- Zhong-Ping Wang
- Clinical Medicine, School of Medicine, Qinghai University, Xining, China
- Department of Respiratory and Critical Diseases, Affiliated Hospital of Qinghai University, Xining, China
| | - Mao Hua
- Department of Respiratory and Critical Diseases, Affiliated Hospital of Qinghai University, Xining, China
| | - Tai Jiu
- Department of Respiratory and Critical Diseases, Affiliated Hospital of Qinghai University, Xining, China
| | - Ri-Li Ge
- Research Center of High-Altitude Medicine, School of Medicine, Qinghai University, Xining, China
- Joint Lab of Qinghai-Utah for High Altitude Medicine, School of Medicine, Qinghai University, Xining, China
| | - Zhenzhong Bai
- Clinical Medicine, School of Medicine, Qinghai University, Xining, China
- Research Center of High-Altitude Medicine, School of Medicine, Qinghai University, Xining, China
- Joint Lab of Qinghai-Utah for High Altitude Medicine, School of Medicine, Qinghai University, Xining, China
| |
Collapse
|
12
|
Raparelli V, Nocella C, Proietti M, Romiti GF, Corica B, Bartimoccia S, Stefanini L, Lenzi A, Viceconte N, Tanzilli G, Cammisotto V, Pilote L, Cangemi R, Basili S, Carnevale R. Testosterone-to-estradiol ratio and platelet thromboxane release in ischemic heart disease: the EVA project. J Endocrinol Invest 2022; 45:1367-1377. [PMID: 35262860 PMCID: PMC9184432 DOI: 10.1007/s40618-022-01771-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/16/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Data on the interplay between sexual hormones balance, platelet function and clinical outcomes of adults with ischemic heart disease (IHD) are still lacking. OBJECTIVE To assess the association between the Testosterone (T)-to-Estradiol (E2) Ratio (T/E2) and platelet activation biomarkers in IHD and its predictive value on adverse outcomes. METHODS The EVA study is a prospective observational study of consecutive hospitalized adults with IHD undergoing coronary angiography and/or percutaneous coronary interventions. Serum T/E2 ratios E2, levels of thromboxane B2 (TxB2) and nitrates (NO), were measured at admission and major adverse events, including all-cause mortality, were collected during a long-term follow-up. RESULTS Among 509 adults with IHD (mean age 67 ± 11 years, 30% females), males were older with a more adverse cluster of cardiovascular risk factors than females. Acute coronary syndrome and non-obstructive coronary artery disease were more prevalent in females versus males. The lower sex-specific T/E2 ratios identified adults with the highest level of serum TxB2 and the lowest NO levels. During a median follow-up of 23.7 months, the lower sex-specific T/E2 was associated with higher all-cause mortality (HR 3.49; 95% CI 1.24-9.80; p = 0.018). In in vitro, platelets incubated with T/E2 ratios comparable to those measured in vivo in the lowest quartile showed increased platelet activation as indicated by higher levels of aggregation and TxB2 production. CONCLUSION Among adults with IHD, higher T/E2 ratio was associated with a lower long-term risk of fatal events. The effect of sex hormones on the platelet thromboxane release may partially explain such finding.
Collapse
Affiliation(s)
- V Raparelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada.
- Department of Translational Medicine, University of Ferrara, via Luigi Borsari, 46, 44121, Ferrara, Italy.
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy.
| | - C Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - M Proietti
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK
| | - G F Romiti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - B Corica
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - S Bartimoccia
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - L Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - N Viceconte
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - G Tanzilli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - V Cammisotto
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, Rome, Italy
| | - L Pilote
- Centre for Outcomes Research and Evaluation, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - R Cangemi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - S Basili
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - R Carnevale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro-Napoli, Naples, Italy
| |
Collapse
|
13
|
Papachristos A, Ratain MJ. Lurbinectedin-induced thrombocytopenia: the role of body surface area. Cancer Chemother Pharmacol 2022; 89:573-575. [PMID: 35362793 PMCID: PMC8972734 DOI: 10.1007/s00280-022-04422-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/11/2022] [Indexed: 11/02/2022]
Abstract
Lurbinectedin is an alkylating agent approved for the second-line treatment of small cell lung cancer. Although initial studies showed no association between body surface area (BSA) and drug clearance, the recommended dose is 3.2 mg/m2 every 3 weeks. This recommendation was based on an exposure-response study, which demonstrated that patients with lower BSA had a higher incidence of thrombocytopenia. Herein we present the factors associated with BSA and thrombopoiesis, which may have contributed to the observed relationship.
Collapse
Affiliation(s)
- Apostolos Papachristos
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, 5841 S. Maryland Ave., MC 2115, Chicago, IL, 60637, USA
| | - Mark J Ratain
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, 5841 S. Maryland Ave., MC 2115, Chicago, IL, 60637, USA. .,Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
14
|
Gérard C, Arnal JF, Jost M, Douxfils J, Lenfant F, Fontaine C, Houtman R, Archer DF, Reid RL, Lobo RA, Gaspard U, Coelingh Bennink HJT, Creinin MD, Foidart JM. Profile of estetrol, a promising native estrogen for oral contraception and the relief of climacteric symptoms of menopause. Expert Rev Clin Pharmacol 2022; 15:121-137. [PMID: 35306927 DOI: 10.1080/17512433.2022.2054413] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Estrogens used in women's healthcare have been associated with increased risks of venous thromboembolism (VTE) and breast cancer. Estetrol (E4), an estrogen produced by the human fetal liver, has recently been approved for the first time as a new estrogenic component of a novel combined oral contraceptive (E4/drospirenone [DRSP]) for over a decade. In phase 3 studies, E4/DRSP showed good contraceptive efficacy, a predictable bleeding pattern, and a favorable safety and tolerability profile. AREAS COVERED This narrative review discusses E4's pharmacological characteristics, mode of action, and the results of preclinical and clinical studies for contraception, as well as for menopause and oncology. EXPERT OPINION Extensive studies have elucidated the properties of E4 that underlie its favorable safety profile. While classical estrogens (such as estradiol) exert their actions via both activation of nuclear and membrane estrogen receptor α (ERα), E4 presents a specific profile of ERα activation: E4 binds and activates nuclear ERα but does not induce the activation of membrane ERα signaling pathways in specific tissues. E4 has a small effect on normal breast tissue proliferation and minimally affects hepatic parameters. This distinct profile of ERα activation, uncoupling nuclear and membrane activation, is unique.
Collapse
Affiliation(s)
- Céline Gérard
- Department Research and Development, Estetra Srl, an Affiliate Company of Mithra Pharmaceuticals, Liège, Belgium
| | - Jean-François Arnal
- CHU de Toulouse, Université Toulouse III, Toulouse, France.,INSERM-UPS UMR U1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Toulouse, France
| | - Maud Jost
- Department Research and Development, Estetra Srl, an Affiliate Company of Mithra Pharmaceuticals, Liège, Belgium
| | - Jonathan Douxfils
- Qualiblood S.a, Namur, Belgium.,Department of Pharmacy, Namur Thrombosis and Hemostasis Center, NAmur Research Institute for Life Sciences, University of Namur, Namur, Belgium
| | - Françoise Lenfant
- CHU de Toulouse, Université Toulouse III, Toulouse, France.,INSERM-UPS UMR U1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Toulouse, France
| | - Coralie Fontaine
- CHU de Toulouse, Université Toulouse III, Toulouse, France.,INSERM-UPS UMR U1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Toulouse, France
| | | | | | - Robert L Reid
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Queen's University, Kingston, Canada
| | - Rogerio A Lobo
- Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, USA
| | - Ulysse Gaspard
- Department of Obstetrics and Gynecology, University of Liège, Liège, Belgium
| | | | - Mitchell D Creinin
- Department of Obstetrics and Gynecology, University of California, Sacramento, USA
| | - Jean-Michel Foidart
- Department Research and Development, Estetra Srl, an Affiliate Company of Mithra Pharmaceuticals, Liège, Belgium.,Department of Obstetrics and Gynecology, University of Liège, Liège, Belgium
| |
Collapse
|
15
|
Cagnacci A, Londero AP, Xholli A. COVID-19 and hormonal contraception. Case Rep Womens Health 2022; 34:e00389. [PMID: 35096531 PMCID: PMC8788094 DOI: 10.1016/j.crwh.2022.e00389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/03/2022] Open
|
16
|
Ma Q, Hao ZW, Wang YF. The effect of estrogen in coronavirus disease 2019. Am J Physiol Lung Cell Mol Physiol 2021; 321:L219-L227. [PMID: 33949212 PMCID: PMC8270516 DOI: 10.1152/ajplung.00332.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
The outcomes of coronavirus disease 2019 (COVID-19) vary between men and women. Some statistical reports have shown that men have a higher risk of developing COVID-19 and suffer from worse outcomes than females. Although there are many factors that can explain the high prevalence of COVID-19 in men, such as lifestyle habits and the different profile of comorbidities among sexes, the distinctions between male and female immune systems cannot be ignored. It has been sufficiently shown that sex differences have a critical influence on the shaping of immune response, which then leads to different pathogenesis in infectious diseases. Compared with males, females typically have a more effective innate and adaptive immune response to viral infections in COVID-19. What's more, there is a growing body of evidence showing that estrogen exerts an effect on the regulation of immune response. This article examines the effect and mechanism of estrogen on COVID-19.
Collapse
Affiliation(s)
- Qing Ma
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhuo-Wen Hao
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan-Feng Wang
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Nayak MK, Ghatge M, Flora GD, Dhanesha N, Jain M, Markan KR, Potthoff MJ, Lentz SR, Chauhan AK. The metabolic enzyme pyruvate kinase M2 regulates platelet function and arterial thrombosis. Blood 2021; 137:1658-1668. [PMID: 33027814 PMCID: PMC7995287 DOI: 10.1182/blood.2020007140] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
Very little is known about the role of metabolic regulatory mechanisms in platelet activation and thrombosis. Dimeric pyruvate kinase M2 (PKM2) is a crucial regulator of aerobic glycolysis that facilitates the production of lactate and metabolic reprogramming. Herein, we report that limiting PKM2 dimer formation, using the small molecule inhibitor ML265, negatively regulates lactate production and glucose uptake in human and murine stimulated platelets. Furthermore, limiting PKM2 dimer formation reduced agonist-induced platelet activation, aggregation, clot retraction, and thrombus formation under arterial shear stress in vitro in both human and murine platelets. Mechanistically, limiting PKM2 dimerization downregulated phosphatidylinositol 3-kinase (PI3K)-mediated protein kinase B or serine/threonine-specific protein kinase (Akt)/glycogen synthase kinase 3 (GSK3) signaling in human and murine platelets. To provide further evidence for the role of PKM2 in platelet function, we generated a megakaryocyte or platelet-specific PKM2-/- mutant strain (PKM2fl/flPF4Cre+). Platelet-specific PKM2-deficient mice exhibited impaired agonist-induced platelet activation, aggregation, clot retraction, and PI3K-mediated Akt/GSK3 signaling and were less susceptible to arterial thrombosis in FeCl3 injury-induced carotid- and laser injury-induced mesenteric artery thrombosis models, without altering hemostasis. Wild-type mice treated with ML265 were less susceptible to arterial thrombosis with unaltered tail bleeding times. These findings reveal a major role for PKM2 in coordinating multiple aspects of platelet function, from metabolism to cellular signaling to thrombosis, and implicate PKM2 as a potential target for antithrombotic therapeutic intervention.
Collapse
Affiliation(s)
- Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Nirav Dhanesha
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Manish Jain
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Kathleen R Markan
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA; and
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA; and
- Department of Veterans Affairs Medical Center, Iowa City, IA
| | - Steven R Lentz
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, and
| |
Collapse
|
18
|
Kumar RS, Goyal N. Estrogens as regulator of hematopoietic stem cell, immune cells and bone biology. Life Sci 2021; 269:119091. [PMID: 33476629 DOI: 10.1016/j.lfs.2021.119091] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells provide continuous supply of all the immune cells, through proliferation and differentiation decisions. These decisions are controlled by local bone marrow environment as well as by long-range signals for example endocrine system. Sex dependent differential immunological responses have been described under homeostasis and disease conditions. Females show higher longevity than male counterpart that seems to depend on major female sex hormone, estrogen. There are four estrogens - Estrone (E1), estradiol (E2), Estriol (E3) and Estetrol (E4) that spatially and temporarily present during different female reproductive phases. In this review, we discussed recent updates describing the effects of estrogen on HSC, immune cells and in bone biology. Estradiol (E2) being a major/abundant estrogen is extensively investigated, while effects of other estrogens E1, E3 and E4 are started to unravel recently. Furthermore, clinical effect of estrogen as hormone therapy is discussed in HSC and immune cells perspectives. The data presented in this review is compiled by searches of PubMed, database of American Cancer Society (ACS). We have included article from September 1994 to March 2020 as covering all article in chronological order is not fissile so we included relevant article with substantial information in this specific area of research by using the search term (alone or in combination) estrogen, hematopoietic stem cell, immune cells, gender difference, estrone, estriol, estetrol, therapeutic application, pregnancy, effect on bone.
Collapse
Affiliation(s)
- Rupali Sani Kumar
- CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India.
| | - Neena Goyal
- CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|
19
|
Abou-Ismail MY, Citla Sridhar D, Nayak L. Estrogen and thrombosis: A bench to bedside review. Thromb Res 2020; 192:40-51. [PMID: 32450447 PMCID: PMC7341440 DOI: 10.1016/j.thromres.2020.05.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/12/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023]
Abstract
Estrogen, in the clinical setting is used primarily for contraception and hormone replacement therapy. It has been well established that estrogen increases the risk of both arterial and venous thrombosis. While estrogen is known to induce a prothrombotic milieu through various effects on the hemostatic pathways, the exact molecular mechanism leading to those effects is not known. The most common clinical presentation of estrogen-related thrombosis is venous thromboembolism (VTE) of the deep veins of the legs or pulmonary vessels, usually within the first few months of use. Estrogen has also been associated with increased risk of "unusual site" thromboses, as well as arterial thrombosis. Women at high-risk of thrombosis need careful evaluation and counseling for contraception, pregnancy, menopausal hormonal therapy and other estrogen-related conditions or treatments in order to lower the risk of thromboses. We review the most recent evidence on management of high-estrogen states in women at high-risk of thrombosis, as well as emerging data on unique populations such as transgender women. More studies are needed to better understand the pathophysiology of hormone-related thrombosis, as well as more comprehensive techniques to stratify risks for thrombosis so as to enable tailoring of recommendations for each individual.
Collapse
Affiliation(s)
- Mouhamed Yazan Abou-Ismail
- Case Western Reserve University, Cleveland, OH, United States of America; University Hospitals Cleveland Medical Center, Cleveland, OH, United States of America
| | - Divyaswathi Citla Sridhar
- Case Western Reserve University, Cleveland, OH, United States of America; Rainbow Babies & Children's Hospital, Cleveland, OH, United States of America
| | - Lalitha Nayak
- Case Western Reserve University, Cleveland, OH, United States of America; University Hospitals Cleveland Medical Center, Cleveland, OH, United States of America.
| |
Collapse
|
20
|
Raparelli V, Proietti M, Lenzi A, Basili S. Sex and Gender Differences in Ischemic Heart Disease: Endocrine Vascular Disease Approach (EVA) Study Design. J Cardiovasc Transl Res 2020; 13:14-25. [PMID: 30511337 PMCID: PMC7010648 DOI: 10.1007/s12265-018-9846-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022]
Abstract
Improvements in ischemic heart disease (IHD) management have been unbalanced between sexes, with coronary microvascular dysfunction considered the likely underlying reason. The Endocrine Vascular disease Approach (EVA) is an observational study (Clinicaltrial.gov NCT02737982) aiming to assess sex and gender interactions between coronary circulation, sexual hormones, and platelet function. Consecutive patients with IHD undergoing coronary angiography will be recruited: (1) to assess sex and gender differences in angiographic reperfusion indexes; (2) to evaluate the effects of estrogen/androgen on sex-related differences in myocardial ischemia; (3) to investigate the platelet biology differences between men and women with IHD; (4) to verify sex- and gender-driven interplay between response to percutaneous coronary intervention, platelets, sex hormones, and myocardial damage at baseline and its impact on 12-month outcomes. The integration of sex and gender in this translational project on IHD will contribute to the identification of new targets for further innovative clinical interventions.
Collapse
Affiliation(s)
- Valeria Raparelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Centre for Outcomes Research and Evaluation, McGill University Health Centre Research Institute, Montreal, QC Canada
| | - Marco Proietti
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
21
|
Breithaupt-Faloppa AC, Correia CDJ, Prado CM, Stilhano RS, Ureshino RP, Moreira LFP. 17β-Estradiol, a potential ally to alleviate SARS-CoV-2 infection. Clinics (Sao Paulo) 2020; 75:e1980. [PMID: 32490931 PMCID: PMC7233687 DOI: 10.6061/clinics/2020/e1980] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
Considering that female sexual hormones may modulate the inflammatory response and also exhibit direct effects on the cells of the immune system, herein, we intend to discuss the sex differences and the role of estradiol in modulating the lung and systemic inflammatory response, focusing on its possible application as a treatment modality for SARS-CoV-2 patients. COVID-19 patients develop severe hypoxemia early in the course of the disease, which is silent most of the time. Small fibrinous thrombi in pulmonary arterioles and a tumefaction of endothelial were observed in the autopsies of fatal COVID-19 cases. Studies showed that the viral infection induces a vascular process in the lung, which included vasodilation and endothelial dysfunction. Further, the proportions of CD4+ T and CD8+ T lymphocytes were strongly reduced in patients with severe SARS-CoV-2 infection. Estradiol is connected with CD4+ T cell numbers and increases T-reg cell populations, affecting immune responses to infection. It is known that estradiol exerts a protective effect on endothelial function, activating the generation of nitric oxide (NO) via endothelial nitric oxide synthase. Estrogen attenuates the vasoconstrictor response to various stimuli and induces vasodilation in the pulmonary vasculature during stress situations like hypoxia. It exerts a variety of rapid actions, which are initiated after its coupling with membrane receptors, which in turn, may positively modulate vascular responses in pulmonary disease and help to maintain microvascular flow. Direct and indirect mechanisms underlying the effects of estradiol were investigated, and the results point to a possible protective effect of estradiol against COVID-19, indicating that it may be considered as an adjuvant therapeutic element for the treatment of patients affected by the novel coronavirus.
Collapse
Affiliation(s)
- Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Cristiano de Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Carla Máximo Prado
- Instituto de Saude e Sociedade (ISS), Universidade Federal de Sao Paulo (UNIFESP), Santos, SP, BR
| | | | - Rodrigo Portes Ureshino
- Departamento de Ciencias Biologicas, Universidade Federal de Sao Paulo (UNIFESP), Diadema, SP, BR
- Laboratorio de Endocrinologia Molecular e Translacional, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
22
|
Coleman JR, Moore EE, Kelher MR, Samuels JM, Cohen MJ, Sauaia A, Banerjee A, Silliman CC, Peltz E. Female platelets have distinct functional activity compared with male platelets: Implications in transfusion practice and treatment of trauma-induced coagulopathy. J Trauma Acute Care Surg 2019; 87:1052-1060. [PMID: 31162329 PMCID: PMC6814522 DOI: 10.1097/ta.0000000000002398] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Females are hypercoagulable and have survival benefit in trauma-induced coagulopathy (TIC). The mechanism for this sex-specific hypercoagulability is unknown. Platelets and platelet function are central in providing hemostatic potential and are the largest contributor to clot strength. Ligands (adenosine diphosphate [ADP] and platelet-activating factor [PAF]) bind distinct platelet receptors to potentiate activation and aggregation. We hypothesize that female platelets have a differential response to ADP and PAF, resulting in greater aggregation and activation compared to males, and that estradiol pretreatment of male or female platelets enhances this activity. METHODS Platelets were collected from healthy volunteers: premenopausal/postmenopausal females (≤54 years, >54 years) and similarly aged males. Platelet aggregometry and flow cytometry (fibrinogen binding capacity) were examined. After treatment with ADP or PAF, platelet aggregation was assessed with Chronolog and activation assessed by CD41 receptor surface expression using flow cytometry. Aggregation and activation were again assessed after platelet pretreatment with estradiol. RESULTS Healthy volunteers included 12 premenopausal and 13 postmenopausal females and 18 similarly aged males. Female platelets (combined premenopausal and postmenopausal) had increased aggregation with ADP stimulation, as compared to male platelets. Male and female platelets had differential fibrinogen receptor expression, with female platelets (combined premenopausal and postmenopausal) demonstrating robust activation with ADP versus male platelets with PAF. In the presence of estradiol incubation, male platelets' activation with PAF approximated that of females (combined premenopausal and postmenopausal) and activation with PAF was enhanced in both male and female platelets. CONCLUSION Male and female platelets have differential response to stimuli, suggesting sex-dependent signaling and cellular activation. Female platelets have both increased aggregation and activation potential, and estradiol pretreatment feminizes male platelets to approximate female platelet activation with PAF. These findings offer potential explanation for sex-based differences in hemostatic potential in TIC and question whether donor sex of transfused platelets should be considered in resuscitation. Estradiol may also serve as a novel therapeutic adjunct in TIC.
Collapse
Affiliation(s)
- Julia R Coleman
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
| | - Ernest E Moore
- Ernest E Moore Shock Trauma Center at Denver Health, Department of Surgery, 777 Bannock St, Denver, CO 80204
| | - Marguerite R Kelher
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
- Vitalant Research Institute-Denver, 717 Yosemite St, Denver, CO 80230
| | - Jason M Samuels
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
| | - Mitchell J Cohen
- Ernest E Moore Shock Trauma Center at Denver Health, Department of Surgery, 777 Bannock St, Denver, CO 80204
| | - Angela Sauaia
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
| | - Anirban Banerjee
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
| | - Christopher C Silliman
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
- Vitalant Research Institute-Denver, 717 Yosemite St, Denver, CO 80230
| | - Erik Peltz
- University of Colorado-Denver, Department of Surgery, 12605 E 16th Ave, Aurora, CO 80045
| |
Collapse
|
23
|
Unsworth AJ, Flora GD, Gibbins JM. Non-genomic effects of nuclear receptors: insights from the anucleate platelet. Cardiovasc Res 2019; 114:645-655. [PMID: 29452349 PMCID: PMC5915957 DOI: 10.1093/cvr/cvy044] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) have the ability to elicit two different kinds of responses, genomic and non-genomic. Although genomic responses control gene expression by influencing the rate of transcription, non-genomic effects occur rapidly and independently of transcriptional regulation. Due to their anucleate nature and mechanistically well-characterized and rapid responses, platelets provide a model system for the study of any non-genomic effects of the NRs. Several NRs have been found to be present in human platelets, and multiple NR agonists have been shown to elicit anti-platelet effects by a variety of mechanisms. The non-genomic functions of NRs vary, including the regulation of kinase and phosphatase activity, ion channel function, intracellular calcium levels, and production of second messengers. Recently, the characterization of mechanisms and identification of novel binding partners of NRs have further strengthened the prospects of developing their ligands into potential therapeutics that offer cardio-protective properties in addition to their other defined genomic effects.
Collapse
Affiliation(s)
- Amanda J Unsworth
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| | - Gagan D Flora
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| | - Jonathan M Gibbins
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| |
Collapse
|
24
|
Dupuis M, Severin S, Noirrit-Esclassan E, Arnal JF, Payrastre B, Valéra MC. Effects of Estrogens on Platelets and Megakaryocytes. Int J Mol Sci 2019; 20:ijms20123111. [PMID: 31242705 PMCID: PMC6627332 DOI: 10.3390/ijms20123111] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/03/2022] Open
Abstract
In women, oral menopausal hormonal therapy (MHT) is associated with adverse effects including an increased incidence of thromboembolic events, classically attributed to an increase in several liver-derived coagulation factors due to hepatic first pass. While platelets are central players in thrombus constitution, their implication in women treated with estrogens remains incompletely characterized. Platelets and their medullar progenitors, megakaryocytes, express estrogen receptors (ER) that may explain, at least in part, a sensitivity to hormonal changes. The purpose of this review is to summarize our current knowledge of estrogen actions on platelets and megakaryocytes in mice following in vivo administration and in women using MHT.
Collapse
Affiliation(s)
- Marion Dupuis
- Inserm, U1048 and Université Toulouse III, I2MC, Toulouse 31432, France.
| | - Sonia Severin
- Inserm, U1048 and Université Toulouse III, I2MC, Toulouse 31432, France.
| | | | | | - Bernard Payrastre
- Inserm, U1048 and Université Toulouse III, I2MC, Toulouse 31432, France.
- CHU de Toulouse, Laboratoire d'Hématologie, Toulouse 31059, France.
| | | |
Collapse
|
25
|
Noirrit E, Buscato M, Dupuis M, Payrastre B, Fontaine C, Arnal JF, Valera MC. Effects of conjugated estrogen and bazedoxifene on hemostasis and thrombosis in mice. Endocr Connect 2019; 8:788-795. [PMID: 31085766 PMCID: PMC6590204 DOI: 10.1530/ec-19-0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 11/09/2022]
Abstract
Estrogen-progestin therapy was previously considered as the standard of care for managing bothersome symptoms associated with menopause, but it increases risks of breast cancer and of thromboembolism. The combination of conjugated estrogen (CE) with bazedoxifene (BZA) named tissue-selective estrogen complex (TSEC) was designed to minimize or even abrogate the undesirable effects on breast, while maintaining the beneficial effects such as prevention of osteoporosis and suppression of climacteric symptoms. The risk on thromboembolism associated with TSEC is unknown, although the clinical available data are reassuring. The aim of this study was to define the impact of a chronic administration of CE, BZA or CE + BZA on hemostasis and thrombosis in ovariectomized mice. As expected, CE, but not BZA neither CE + BZA, induced uterine and vagina hypertrophy. As previously demonstrated for 17β-estradiol (E2), we found that CE (i) increased tail-bleeding time, (ii) prevented occlusive thrombus formation in injured carotid artery and (iii) protected against collagen/epinephrine-induced thromboembolism. Thus, whereas BZA antagonized CE action on reproductive tissues, it had no impact on the effect of CE on hemostasis, thromboembolism and arterial thrombosis in mice. CE + BZA shared the anti-thrombotic actions of CE in these mouse models. If a similar process is at work in women, CE combined with BZA could contribute to minimize the risk of thrombosis associated with hormone replacement therapy.
Collapse
Affiliation(s)
- Emmanuelle Noirrit
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
- Faculté de Chirurgie Dentaire, Université de Toulouse III, Toulouse, France
| | - Mélissa Buscato
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
| | - Marion Dupuis
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
| | - Bernard Payrastre
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
| | - Coralie Fontaine
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
| | - Jean-François Arnal
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
| | - Marie-Cécile Valera
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, I2MC, Toulouse, France
- Faculté de Chirurgie Dentaire, Université de Toulouse III, Toulouse, France
- Correspondence should be addressed to M-C Valera:
| |
Collapse
|
26
|
Valéra MC, Noirrit-Esclassan E, Dupuis M, Fontaine C, Lenfant F, Briaux A, Cabou C, Garcia C, Lairez O, Foidart JM, Payrastre B, Arnal JF. Effect of estetrol, a selective nuclear estrogen receptor modulator, in mouse models of arterial and venous thrombosis. Mol Cell Endocrinol 2018; 477:132-139. [PMID: 29928930 DOI: 10.1016/j.mce.2018.06.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/13/2018] [Accepted: 06/16/2018] [Indexed: 01/27/2023]
Abstract
Estetrol (E4) is a natural estrogen synthesized exclusively during pregnancy by the human fetal liver, and the physiological role of this hormone is unknown. Interestingly, E4 was recently evaluated in preclinical and phase II-III clinical studies in combination with a progestin, with the advantage to not increase the circulating level of coagulation factors, at variance to oral estradiol or ethinylestradiol. Here, we evaluated the effect of E4 on hemostasis and thrombosis in mouse. Following chronic E4 treatment, mice exhibited a prolonged tail-bleeding time and were protected from arterial and also venous thrombosis in vivo. In addition, E4 treatment decreased ex vivo thrombus growth on collagen under arterial flow conditions. We recently showed that E4 activates uterine epithelial proliferation through nuclear estrogen receptor (ER) α. To analyze the impact of nuclear ERα actions on hemostasis and thrombosis, we generated hematopoietic chimera with bone marrow cells deficient for nuclear ERα. E4-induced protection against thromboembolism was significantly reduced in the absence of hematopoietic nuclear ERα activation, while the increased tail-bleeding time was not impacted by this deletion. In addition to its "liver friendly" profile described in women, our data shows that E4 has anti-thrombotic properties in various mouse models. Altogether, the natural fetal estrogen E4 could represent an attractive alternative to classic estrogens in oral contraception and treatment of menopause.
Collapse
Affiliation(s)
- Marie-Cécile Valéra
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France; Faculté de Chirurgie Dentaire, Université de Toulouse III, Toulouse, France
| | - Emmanuelle Noirrit-Esclassan
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France; Faculté de Chirurgie Dentaire, Université de Toulouse III, Toulouse, France
| | - Marion Dupuis
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Coralie Fontaine
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Françoise Lenfant
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Anne Briaux
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Cendrine Cabou
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Cedric Garcia
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - Olivier Lairez
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France
| | - Jean-Michel Foidart
- Laboratory of Tumor and Development Biology GIGA-Cancer, Institute of Pathology, University of Liège, CHU-B23, B-4000, Liège, Belgium
| | - Bernard Payrastre
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France; Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Inserm U1048, CHU de Toulouse and Université de ToulouseToulouse, France.
| |
Collapse
|
27
|
Piel-Julian ML, Mahévas M, Germain J, Languille L, Comont T, Lapeyre-Mestre M, Payrastre B, Beyne-Rauzy O, Michel M, Godeau B, Adoue D, Moulis G. Risk factors for bleeding, including platelet count threshold, in newly diagnosed immune thrombocytopenia adults. J Thromb Haemost 2018; 16:1830-1842. [PMID: 29978544 DOI: 10.1111/jth.14227] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/23/2018] [Indexed: 01/19/2023]
Abstract
Essentials Risk factors of bleeding in adult immune thrombocytopenia are not known. This multicenter study assessed risk factors of bleeding at immune thrombocytopenia onset. Platelet count thresholds associated with bleeding were < 20 × 109 L-1 and < 10 × 109 L-1 . Exposure to anticoagulants was a major risk factor of severe bleeding. SUMMARY Background The aim of this cross-sectional study was to assess risk factors for bleeding in immune thrombocytopenia (ITP) adults, including the determination of platelet count thresholds. Methods We selected all newly diagnosed ITP adults included in the Cytopénies Auto-immunes Registre Midi-PyrénéEN (CARMEN) register and at the French referral center for autoimmune cytopenias. The frequencies of any bleeding, mucosal bleeding and severe bleeding (gastrointestinal, intracranial, or macroscopic hematuria) at ITP onset were assessed. Platelet count thresholds were assessed by the use of receiver operating characteristic curves. All potential risk factors were included in logistic regression models. Results Among the 302 patients, the frequencies of any, mucosal and severe bleeding were 57.9%, 30.1%, and 6.6%, respectively. The best discriminant threshold of platelet count for any bleeding was 20 × 109 L-1 . In multivariate analysis, factors associated with any bleeding were platelet count (< 10 × 109 L-1 versus ≥ 20 × 109 L-1 , odds ratio [OR] 48.2, 95% confidence interval [CI] 20.0-116.3; between 10 × 109 L-1 and 19 × 109 L-1 versus ≥ 20 × 109 L-1 , OR 5.2, 95% CI 2.3-11.6), female sex (OR 2.6, 95% CI 1.3-5.0), and exposure to non-steroidal anti-inflammatory drugs (NSAIDs) (OR 4.8, 95% CI 1.1-20.7). A low platelet count was also the main risk factor for mucosal bleeding. Exposure to anticoagulant drugs was associated with severe bleeding (OR 4.3, 95% CI 1.3-14.1). Conclusions Platelet counts of < 20 × 109 L-1 and < 10 × 109 L-1 were thresholds for major increased risks of any and mucosal bleeding. Platelet count, female sex and exposure to NSAIDs should be considered for assessment of the risk of any bleeding. Exposure to anticoagulant drugs was a major risk factor for severe bleeding.
Collapse
Affiliation(s)
- M-L Piel-Julian
- Service de Médecine Interne, Salle Le Tallec, Centre Hospitalier Universitaire de Toulouse-Purpan, Toulouse, France
| | - M Mahévas
- Service de Médecine Interne, CHU Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Cytopénies Auto-Immunes de l'adulte, Université Paris-Est-Créteil, Créteil, France
| | - J Germain
- Centre d'Investigation Clinique 1436, Axe Pharmacoépidémiologie, Centre Hospitalier Universitaire de Toulouse-Purpan, Toulouse, France
| | - L Languille
- Service de Médecine Interne, CHU Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Cytopénies Auto-Immunes de l'adulte, Université Paris-Est-Créteil, Créteil, France
| | - T Comont
- Service de Médecine Interne, Institut Universitaire du Cancer de Toulouse - Oncopôle, Toulouse, France
| | - M Lapeyre-Mestre
- Centre d'Investigation Clinique 1436, Axe Pharmacoépidémiologie, Centre Hospitalier Universitaire de Toulouse-Purpan, Toulouse, France
- UMR 1027, INSERM-Université de Toulouse, Toulouse, France
- Service de Pharmacologie Médicale et Clinique, CHU de Toulouse, Toulouse, France
| | - B Payrastre
- Inserm U1048 (I2MC), CHU de Toulouse and Université Toulouse III, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - O Beyne-Rauzy
- Service de Médecine Interne, Institut Universitaire du Cancer de Toulouse - Oncopôle, Toulouse, France
| | - M Michel
- Service de Médecine Interne, CHU Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Cytopénies Auto-Immunes de l'adulte, Université Paris-Est-Créteil, Créteil, France
| | - B Godeau
- Service de Médecine Interne, CHU Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Cytopénies Auto-Immunes de l'adulte, Université Paris-Est-Créteil, Créteil, France
| | - D Adoue
- Service de Médecine Interne, Institut Universitaire du Cancer de Toulouse - Oncopôle, Toulouse, France
| | - G Moulis
- Service de Médecine Interne, Salle Le Tallec, Centre Hospitalier Universitaire de Toulouse-Purpan, Toulouse, France
- Centre d'Investigation Clinique 1436, Axe Pharmacoépidémiologie, Centre Hospitalier Universitaire de Toulouse-Purpan, Toulouse, France
- UMR 1027, INSERM-Université de Toulouse, Toulouse, France
| |
Collapse
|
28
|
Vinel A, Coudert AE, Buscato M, Valera MC, Ostertag A, Katzenellenbogen JA, Katzenellenbogen BS, Berdal A, Babajko S, Arnal JF, Fontaine C. Respective role of membrane and nuclear estrogen receptor (ER) α in the mandible of growing mice: Implications for ERα modulation. J Bone Miner Res 2018; 33:1520-1531. [PMID: 29624728 PMCID: PMC6563159 DOI: 10.1002/jbmr.3434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/21/2018] [Accepted: 03/27/2018] [Indexed: 12/17/2022]
Abstract
Estrogens play an important role in bone growth and maturation as well as in the regulation of bone turnover in adults. Although the effects of 17β-estradiol (E2) are well documented in long bones and vertebrae, little is known regarding its action in the mandible. E2 actions could be mediated by estrogen receptor (ER) α or β. ERs act primarily as transcriptional factors through two activation functions (AFs), AF1 and AF2, but they can also elicit membrane-initiated steroid signaling (MISS). The aim of the present study was to define ER pathways involved in E2 effects on mandibular bone. Using mice models targeting ERβ or ERα, we first show that E2 effects on mandibular bone are mediated by ERα and do not require ERβ. Second, we show that nuclear ERαAF2 is absolutely required for all the actions of E2 on mandibular bone. Third, inactivation of ERαMISS partially reduced the E2 response on bone thickness and volume, whereas there was no significant impact on bone mineral density. Altogether, these results show that both nuclear and membrane ERα are requested to mediate full estrogen effects in the mandible of growing mice. Finally, selective activation of ERαMISS is able to exert an effect on alveolar bone but not on the cortical compartment, contrary to its protective action on femoral cortical bone. To conclude, these results highlight similarities but also specificities between effects of estrogen in long bones and in the mandible that could be of interest in therapeutic approaches to treat bone mass reduction. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alexia Vinel
- INSERM-U 1048, I2MC, University of Toulouse 3, Toulouse, France
| | - Amelie E Coudert
- Molecular Oral Pathophysiology Team, Centre de Recherche des Cordeliers, INSERM-U 1138, University of Paris-Diderot, Paris, France
| | - Melissa Buscato
- INSERM-U 1048, I2MC, University of Toulouse 3, Toulouse, France
| | | | - Agnès Ostertag
- UMR1132, BIOSCAR, University of Paris-Diderot, Paris, France
| | | | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ariane Berdal
- Molecular Oral Pathophysiology Team, Centre de Recherche des Cordeliers, INSERM-U 1138, University of Paris-Diderot, Paris, France
| | - Sylvie Babajko
- Molecular Oral Pathophysiology Team, Centre de Recherche des Cordeliers, INSERM-U 1138, University of Paris-Diderot, Paris, France
| | | | | |
Collapse
|
29
|
Farris M, Bastianelli C, Rosato E, Brosens I, Benagiano G. Pharmacodynamics of combined estrogen-progestin oral contraceptives: 2. effects on hemostasis. Expert Rev Clin Pharmacol 2017; 10:1129-1144. [DOI: 10.1080/17512433.2017.1356718] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Manuela Farris
- Department of Gynecology, Obstetrics and Urology, Sapienza, University of Rome, Rome, Italy
- AIED, Rome, Italy
| | - Carlo Bastianelli
- Department of Gynecology, Obstetrics and Urology, Sapienza, University of Rome, Rome, Italy
| | - Elena Rosato
- Department of Gynecology, Obstetrics and Urology, Sapienza, University of Rome, Rome, Italy
| | - Ivo Brosens
- LIFE, Leuven Institute for Fertility and Embryology, Leuven, Belgium
| | - Giuseppe Benagiano
- Department of Gynecology, Obstetrics and Urology, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
30
|
Schoeman RM, Lehmann M, Neeves KB. Flow chamber and microfluidic approaches for measuring thrombus formation in genetic bleeding disorders. Platelets 2017; 28:463-471. [PMID: 28532218 PMCID: PMC6131111 DOI: 10.1080/09537104.2017.1306042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Platelet adhesion and aggregation, coagulation, fibrin formation, and fibrinolysis are regulated by the forces and flows imposed by blood at the site of a vascular injury. Flow chambers designed to observe these events are an indispensable part of doing hemostasis and thrombosis research, especially with human blood. Microfluidic methods have provided the flexibility to design flow chambers with complex geometries and features that more closely mimic the anatomy and physiology of blood vessels. Additionally, microfluidic systems with integrated optics and/or pressure sensors and on-board signal processing could transform what have been primarily research tools into clinical assays. Here, we describe a historical review of how flow-based approaches have informed biophysical mechanisms in genetic bleeding disorders, challenges and potential solutions for developing models of bleeding in vitro, and outstanding issues that need to be addressed prior to their use in clinical settings.
Collapse
Affiliation(s)
- Rogier M. Schoeman
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, CO, USA
| | - Marcus Lehmann
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, CO, USA
| | - Keith B. Neeves
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, CO, USA
- Pediatrics, University of Colorado, Denver, CO, USA
| |
Collapse
|
31
|
Effect of chronic estradiol plus progesterone treatment on experimental arterial and venous thrombosis in mouse. PLoS One 2017; 12:e0177043. [PMID: 28486478 PMCID: PMC5423617 DOI: 10.1371/journal.pone.0177043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/23/2017] [Indexed: 11/27/2022] Open
Abstract
Postmenopausal hormone replacement therapy (HRT) with estrogen plus progestogens is the first line therapy to treat menopausal symptoms. The progestogen is added to estrogen to reduce or eliminate the excess risk of endometrial cancer due to the unopposed effect of estrogen. Whereas progestin clearly opposes the proliferative and deleterious long-term actions of estrogen on the endometrium, the interference of progestin on the other estrogen action remains unclear. We previously reported that chronic subcutaneous 17α-estradiol (E2) in mice decreases platelet responsiveness, prolongs the tail-bleeding time and protects against acute thromboembolism. Here, we report the tissue-specific interference of progesterone (P4) on the action of E2 in ovariectomized mice. We first confirm that, in our experimental conditions, P4 attenuates the proliferative action of E2 on the uterus and the effects of E2 on vagina weight and lubrication. We then studied the effect of E2 combined with P4 on hemostasis and thrombosis in vivo in mice and found that P4 did not interfere with the main actions of E2 on platelets, bleeding time and arterial and venous thrombosis. Thus, whereas activation of progesterone receptor interferes with the action of E2 on its classic sex targets, P4 appears to have minimal effect on the hemostasis and thrombosis actions of E2, supporting the prominent role of estrogens and the accessory role of natural progestin on the extra-reproductive cells and tissues involved in thrombosis.
Collapse
|
32
|
Gigante A, Di Mario F, Pierucci A, Amoroso A, Pignataro FS, Napoleone L, Basili S, Raparelli V. Kidney disease and venous thromboembolism: Does being woman make the difference? Eur J Intern Med 2017; 39:18-23. [PMID: 28258791 DOI: 10.1016/j.ejim.2017.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/20/2016] [Accepted: 02/16/2017] [Indexed: 01/05/2023]
Abstract
The risk of venous thromboembolism (VTE) is increased across the spectrum of chronic kidney disease (CKD), from mild to more advanced CKD, and typically characterizes nephrotic syndrome (NS). VTE risk in patients with kidney disease may be due to underlying hemostatic abnormalities, including activation of pro-thrombotic factors, inhibition of endogenous anticoagulation systems, enhanced platelet activation and aggregation, and decreased fibrinolytic activity. The mechanisms involved differ depending on the cause of the kidney impairment (i.e. presence of NS or CKD stage). Sex and gender differences, as well as, environmental factors or comorbidities may play a modulating role; however, specific sex and gender data on this topic are still rare. The aim of the present review is to discuss the VTE risk associated with impairment of kidney function, the potential mechanism accounting for it and the impact of sex differences in this clinical setting.
Collapse
Affiliation(s)
- A Gigante
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy.
| | - F Di Mario
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - A Pierucci
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - A Amoroso
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - F S Pignataro
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - L Napoleone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - S Basili
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy; Research Center on Gender and Evaluation and Promotion of Quality in Medicine, (CEQUAM), Sapienza University of Rome, Rome, Italy
| | - V Raparelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Shi X, Yang J, Cui X, Huang J, Long Z, Zhou Y, Liu P, Tao L, Ruan Z, Xiao B, Zhang W, Li D, Dai K, Mao J, Xi X. Functional Effect of the Mutations Similar to the Cleavage during Platelet Activation at Integrin β3 Cytoplasmic Tail when Expressed in Mouse Platelets. PLoS One 2016; 11:e0166136. [PMID: 27851790 PMCID: PMC5112943 DOI: 10.1371/journal.pone.0166136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022] Open
Abstract
Previous studies in Chinese hamster ovary cells showed that truncational mutations of β3 at sites of F754 and Y759 mimicking calpain cleavage regulate integrin signaling. The roles of the sequence from F754 to C-terminus and the conservative N756ITY759 motif in platelet function have yet to be elaborated. Mice expressing β3 with F754 and Y759 truncations, or NITY deletion (β3-ΔTNITYRGT, β3-ΔRGT, or β3-ΔNITY) were established through transplanting the homozygous β3-deficient mouse bone marrow cells infected by the GFP tagged MSCV MigR1 retroviral vector encoding different β3 mutants into lethally radiated wild-type mice. The platelets were harvested for soluble fibrinogen binding and platelet spreading on immobilized fibrinogen. Platelet adhesion on fibrinogen- and collagen-coated surface under flow was also tested to assess the ability of the platelets to resist hydrodynamic drag forces. Data showed a drastic inhibition of the β3-ΔTNITYRGT platelets to bind soluble fibrinogen and spread on immobilized fibrinogen in contrast to a partially impaired fibrinogen binding and an almost unaffected spreading exhibited in the β3-ΔNITY platelets. Behaviors of the β3-ΔRGT platelets were consistent with the previous observations in the β3-ΔRGT knock-in platelets. The adhesion impairment of platelets with the β3 mutants under flow was in different orders of magnitude shown as: β3-ΔTNITYRGT>β3-ΔRGT>β3-ΔNITY to fibrinogen-coated surface, and β3-ΔTNITYRGT>β3-ΔNITY>β3-ΔRGT to collagen-coated surface. To evaluate the interaction of the β3 mutants with signaling molecules, GST pull-down and immunofluorescent assays were performed. Results showed that β3-ΔRGT interacted with kindlin but not c-Src, β3-ΔNITY interacted with c-Src but not kindlin, while β3-ΔTNITYRGT did not interact with both proteins. This study provided evidence in platelets at both static and flow conditions that the calpain cleavage-related sequences of integrin β3, i.e. T755NITYRGT762, R760GT762, and N756ITY759 participate in bidirectional, outside-in, and inside-out signaling, respectively and the association of c-Src or kindlin with β3 integrin may regulate these processes.
Collapse
Affiliation(s)
- Xiaofeng Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jichun Yang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiongying Cui
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiansong Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Hematology, Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhangbiao Long
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yulan Zhou
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ping Liu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lanlan Tao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng Ruan
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bing Xiao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Dongya Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, 215006, China
| | - Jianhua Mao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- * E-mail: (JM); (XX)
| | - Xiaodong Xi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- * E-mail: (JM); (XX)
| |
Collapse
|
34
|
Vinel A, Hay E, Valera MC, Buscato M, Adlanmerini M, Guillaume M, Cohen-Solal M, Ohlsson C, Lenfant F, Arnal JF, Fontaine C. Role of ERαMISS in the Effect of Estradiol on Cancellous and Cortical Femoral Bone in Growing Female Mice. Endocrinology 2016; 157:2533-44. [PMID: 27105385 DOI: 10.1210/en.2015-1994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Estrogen receptor-α (ERα) acts primarily in the nucleus as a transcription factor involving two activation functions, AF1 and AF2, but it can also induce membrane-initiated steroid signaling (MISS) through the modulation of various kinase activities and/or secondary messenger levels. Previous work has demonstrated that nuclear ERα is required for the protective effect of the estrogen 17β-estradiol (E2), whereas the selective activation of ERαMISS is sufficient to confer protection in cortical but not cancellous bone. The aim of this study was to define whether ERαMISS is necessary for the beneficial actions of chronic E2 exposure on bone. We used a mouse model in which ERα membrane localization had been abrogated due to a point mutation of the palmitoylation site of ERα (ERα-C451A). Alterations of the sex hormones in ERα-C451A precluded the interpretation of bone parameters that were thus analyzed on ovariectomized and supplemented or not with E2 (8 μg/kg/d) to circumvent this bias. We found the beneficial action of E2 on femoral bone mineral density as well as in both cortical and cancellous bone was decreased in ERα-C451A mice compared with their wild-type littermates. Histological and biochemical approaches concurred with the results from bone marrow chimeras to demonstrate that ERαMISS signaling affects the osteoblast but not the osteoclast lineage in response to E2. Thus, in contrast to the uterine and endothelial effects of E2 that are specifically mediated by nuclear ERα and ERαMISS effects, respectively, bone protection is dependent on both, underlining the exquisite tissue-specific actions and interactions of membrane and nuclear ERα.
Collapse
Affiliation(s)
- Alexia Vinel
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Eric Hay
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Marie-Cécile Valera
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Mélissa Buscato
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Marine Adlanmerini
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Maeva Guillaume
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Martine Cohen-Solal
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Claes Ohlsson
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Françoise Lenfant
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Jean-François Arnal
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Coralie Fontaine
- INSERM Unité 1048 (A.V., M.C.V., M.B., M.A., M.G., F.L., J.F.A., C.F.), I2MC, University of Toulouse 3, F-31432 Toulouse, France; Unité Mixte de Recherche 1132 (E.H., M.C.-S.), Bone and Cartilage Biology, University of Paris 7, F-75006 Paris, France; and Centre for Bone and Arthritis Research (C.O.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| |
Collapse
|
35
|
Palierne G, Fabre A, Solinhac R, Le Péron C, Avner S, Lenfant F, Fontaine C, Salbert G, Flouriot G, Arnal JF, Métivier R. Changes in Gene Expression and Estrogen Receptor Cistrome in Mouse Liver Upon Acute E2 Treatment. Mol Endocrinol 2016; 30:709-32. [PMID: 27164166 DOI: 10.1210/me.2015-1311] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Transcriptional regulation by the estrogen receptor-α (ER) has been investigated mainly in breast cancer cell lines, but estrogens such as 17β-estradiol (E2) exert numerous extrareproductive effects, particularly in the liver, where E2 exhibits both protective metabolic and deleterious thrombotic actions. To analyze the direct and early transcriptional effects of estrogens in the liver, we determined the E2-sensitive transcriptome and ER cistrome in mice after acute administration of E2 or placebo. These analyses revealed the early induction of genes involved in lipid metabolism, which fits with the crucial role of ER in the prevention of liver steatosis. Characterization of the chromatin state of ER binding sites (BSs) in mice expressing or not ER demonstrated that ER is not required per se for the establishment and/or maintenance of chromatin modifications at the majority of its BSs. This is presumably a consequence of a strong overlap between ER and hepatocyte nuclear factor 4α BSs. In contrast, 40% of the BSs of the pioneer factor forkhead box protein a (Foxa2) were dependent upon ER expression, and ER expression also affected the distribution of nucleosomes harboring dimethylated lysine 4 of Histone H3 around Foxa2 BSs. We finally show that, in addition to a network of liver-specific transcription factors including CCAAT/enhancer-binding protein and hepatocyte nuclear factor 4α, ER might be required for proper Foxa2 function in this tissue.
Collapse
Affiliation(s)
- Gaëlle Palierne
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Aurélie Fabre
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Romain Solinhac
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Christine Le Péron
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Stéphane Avner
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Françoise Lenfant
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Coralie Fontaine
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Gilles Salbert
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Gilles Flouriot
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Jean-François Arnal
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| | - Raphaël Métivier
- Equipe Spatio-Temporal Regulation of Transcription in Eukaryotes (SP@RTE) (G.P., C.L.P., S.A., G.S., R.M.), Unité Mixte de Recherche 6290 Centre National de la Recherche Scientifique (Institut de Genétique et Développement de Rennes), Université de Rennes 1, Campus de Beaulieu, and Equipe Transcription, Environment and Cancer (TREC) (G.F.), Inserm U1085-Institut de Recherche en Santé, Environnement et Travail, Rennes 35042 Cedex, France; and Equipe 9 "Estrogen Receptor: In Vivo Dissection and Modulation" (A.F., R.S., F.L., C.F., J.-F.A.), Inserm Unité 1048 (Institut des Maladies Métaboliques et Cardiovasculaires), Toulouse 31432 Cedex 4, France
| |
Collapse
|
36
|
Shi X, Yang J, Huang J, Long Z, Ruan Z, Xiao B, Xi X. Effects of different shear rates on the attachment and detachment of platelet thrombi. Mol Med Rep 2016; 13:2447-56. [PMID: 26847168 PMCID: PMC4768970 DOI: 10.3892/mmr.2016.4825] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 01/15/2016] [Indexed: 11/06/2022] Open
Abstract
Thrombosis and hemostasis take place in flowing blood, which generates shear forces. The effect of different shear rates, particularly pathological forces, on platelet thrombus formation remains to be fully elucidated. The present study observed the morphological characteristics and hierarchical structure of thrombi on the collagen surface at a wide range of wall shear rates (WSRs) and examined the underlying mechanisms. Calcein AM‑labeled whole blood was perfused over a collagen‑coated surface at different shear rates set by a Bioflux 200 microfluidic device and the thrombi formed were assessed for area coverage, the height and the hierarchical structure defined by the extent of platelet activation and packing density. The factors that affect thrombus formation were also investigated. Platelet thrombus formation varied under different WSRs, for example, dispersed platelet adhesion mixed with erythrocytes was observed at 125‑250 s(‑1), extensive and thin platelet thrombi were observed at 500‑1,500 s(‑1), and sporadic, thick thrombi were observed at pathological WSRs of 2,500‑5,000 s(‑1), which showed a tendency to be shed. With increasing WSRs, the height of the thrombi showed an increasing linear trend, whereas the total fluorescence intensity and area of the thrombi exhibited a parabolic curve‑like change, with a turning point at a WSR of 2,500 s(‑1). The number of thrombi, the average fluorescence intensity and the area per thrombus showed similar trends, with an initial upwards incline followed by a decline. The thrombi formed at higher WSRs had a thicker shell, which led to a more densely packed core. Platelet thrombus formation under shear‑flow was regulated by the adhesive strength, which was mediated by receptor‑ligand interaction, the platelet deposition induced by shear rates and the detachment by the dynamic force of flow. This resulted in a balance between thrombus attachment, including adhesion and aggregation, and detachment. Collectively, compared with physiological low WSRs, pathological high WSRs caused thicker and more easily shed thrombi with more condensed cores, which was regulated by an attachment‑detachment balance. These results provide novel insights into the properties of thrombus formation on collagen at different WSRs, and offers possible explanations for certain clinical physiopathological phenomena, including physical hemostasis and pathological thrombosis.
Collapse
Affiliation(s)
- Xiaofeng Shi
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Jichun Yang
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Jiansong Huang
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Zhangbiao Long
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Zheng Ruan
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Bing Xiao
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Xiaodong Xi
- Department of Hematology, Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Sino‑French Research Center for Life Sciences and Genomics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| |
Collapse
|
37
|
Komsa-Penkova R, Golemanov G, Tsankov B, Ivanov P, Beshev L, Tonchev P. Rs5918ITGB3 Polymorphism, Smoking, and BMI as Risk Factors for Early Onset and Recurrence of DVT in Young Women. Clin Appl Thromb Hemost 2016; 23:585-595. [DOI: 10.1177/1076029615624778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective: To evaluate the contribution of rs5918ITGB3 on the incidence and recurrence of deep venous thrombosis (DVT) in women and the relationship with body mass index (BMI) and smoking and to compare with data in men. Results: Rs5918(C) polymorphism in ITGB3 gene was assessed in 224 patients diagnosed with DVT and 216 controls. Thrombophilic genetic variant rs5918(C) was significantly pronounced in women (χ2 =7.565, P = .008) and total patients (χ2 = 9.266, P = .002) but not in men. Women patients (<45 years) who were carriers of rs5918ITGB3 polymorphism had an early onset of DVT (34.5 vs 39.4 years, χ2 = 7.027, P = .008) as analyzed by Kaplan-Meier and a higher risk of the recurrent event (χ2 = 3.405, odds ratio = 2.581, P = .044). The period before recurrent venous thromboembolism event was related to smoking status and BMI in young female who were carriers of rs5918 polymorphism but not in the males. Conclusions: Carriage of genetic variant rs5918(C) polymorphism in ITGB3 gene in women contributes to higher risk of single and recurrent DVT events at younger age.
Collapse
Affiliation(s)
| | - Georgi Golemanov
- Department of Biochemistry, Medical University-Pleven, Pleven, Bulgaria
| | - Boris Tsankov
- Department of Surgery, University Hospital, Medical University-Pleven, Pleven, Bulgaria
| | - Petar Ivanov
- Department of Biochemistry, Medical University-Pleven, Pleven, Bulgaria
| | - Lyubomir Beshev
- Department of Surgery, University Hospital, Medical University-Pleven, Pleven, Bulgaria
| | - Pencho Tonchev
- Department of Surgery, University Hospital, Medical University-Pleven, Pleven, Bulgaria
| |
Collapse
|
38
|
Valéra MC, Fontaine C, Lenfant F, Cabou C, Guillaume M, Smirnova N, Kim SH, Chambon P, Katzenellenbogen JA, Katzenellenbogen BS, Payrastre B, Arnal JF. Protective Hematopoietic Effect of Estrogens in a Mouse Model of Thrombosis: Respective Roles of Nuclear Versus Membrane Estrogen Receptor α. Endocrinology 2015; 156:4293-301. [PMID: 26280130 PMCID: PMC4606746 DOI: 10.1210/en.2015-1522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We recently reported that chronic 17β-estradiol (E2) treatment in mice decreases platelet responsiveness, prolongs the tail-bleeding time and protects against acute thromboembolism via the hematopoietic estrogen receptor alpha (ERα), and independently of ERβ. Here, we have explored the respective roles of membrane vs nuclear actions of ERα in this process, using: 1) the selective activator of membrane ERα: estrogen dendrimer conjugate, and 2) mouse models with mutations in ERα. The selective targeting of activation function 2 of ERα provides a model of nuclear ERα loss-of-function, whereas mutation of the ERα palmitoylation site leads to a model of membrane ERα deficiency. The combination of pharmacological and genetic approaches including hematopoietic chimera mice demonstrated that absence of either membrane or nuclear ERα activation in bone marrow does not prevent the prolongation of the tail-bleeding time, suggesting a redundancy of these two functions for this E2 effect. In addition, although hematopoietic membrane ERα is neither sufficient nor necessary to protect E2-treated mice from collagen/epinephrine-induced thromboembolism, the protection against death-induced thromboembolism is significantly reduced in the absence of hematopoietic nuclear ERα activation. Overall, this study emphasizes that hematopoietic cells (likely megakaryocytes and possibly immune cells) constitute an important target in the antithrombotic effects of estrogens, and delineate for the first time in vivo the respective roles of membrane vs nuclear ERα effects, with a prominent role of the latter.
Collapse
Affiliation(s)
- Marie-Cécile Valéra
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Coralie Fontaine
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Françoise Lenfant
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Cendrine Cabou
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Maeva Guillaume
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Natalia Smirnova
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Sung Hoon Kim
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Pierre Chambon
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - John A Katzenellenbogen
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Benita S Katzenellenbogen
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Bernard Payrastre
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| | - Jean-François Arnal
- INSERM U1048 and Université Toulouse III I2MC (M.-C.V., C.F., F.L., C.C., M.G., N.S., B.P., J.-F.A.), Toulouse, 31432 France; Faculté de Chirurgie Dentaire (M.-C.V.), Université de Toulouse III, Toulouse, 31432 France; Department of Chemistry (H.H.K., J.A.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), Collège de France, Université de Strasbourg, Illkirch, 67400 France; Department of Physiology and Cell Biology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, IL 61801; and Laboratoire d'Hématologie (B.P.), Centre Hospitalier Universitaire de Toulouse, Toulouse, 31432 France
| |
Collapse
|
39
|
Valera MC, Gourdy P, Trémollières F, Arnal JF. From the Women’s Health Initiative to the combination of estrogen and selective estrogen receptor modulators to avoid progestin addition. Maturitas 2015; 82:274-7. [DOI: 10.1016/j.maturitas.2015.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/27/2022]
|
40
|
Valéra MC, Parant O, Cenac C, Arnaud C, Gallini A, Hamdi S, Sié P, Arnal JF, Payrastre B. Platelet Adhesion and Thrombus Formation in Whole Blood at Arterial Shear Rate at the End of Pregnancy. Am J Reprod Immunol 2015; 74:533-41. [PMID: 26435170 DOI: 10.1111/aji.12433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 09/09/2015] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Platelet reactivity has not been evaluated in integrated functional testing during normal pregnancy. Here, we analysed platelet functions under arterial shear rate in comparison with static conditions. METHOD OF STUDY Thirty pregnant women with uncomplicated pregnancies and 30 healthy non-pregnant women were enrolled in this study. Platelet adhesion to collagen and fibrinogen and subsequent thrombus formation were measured at arterial shear rate in whole blood using a microfluidic and imaging system. Standard light transmission aggregometry, flow cytometry of activation markers in washed platelets and impedance aggregometry in whole blood were also used to assess platelet responsiveness in static conditions. RESULTS Compared to non-pregnant controls, thrombus formation on collagen fibres and firm platelet adhesion on fibrinogen under arterial shear rate were significantly reduced in pregnant women. Platelet aggregometry assays in suspension showed a slight increase in platelet reactivity in pregnant women. CONCLUSION While platelet aggregometry and platelet activation markers in static conditions show little changes in platelet reactivity, monitoring of platelet adhesion and thrombus growth on collagen or fibrinogen under flow condition in whole blood indicates a significant decrease in pregnant women compared to controls. This decrease might contribute to counteract a hypercoagulable state and to reduce the risk of arterial thrombosis.
Collapse
Affiliation(s)
- Marie-Cécile Valéra
- INSERM, U1048, CHU de Toulouse and Université Toulouse 3, Toulouse Cedex 04, France
| | - Olivier Parant
- Service de Gynécologie Obstétrique, CHU de Toulouse, Toulouse, France
| | - Claire Cenac
- INSERM, U1043, CNRS, U5282 and Université Toulouse 3, CPTP, Toulouse, France
| | - Catherine Arnaud
- INSERM, U1027 and Université Toulouse 3, Toulouse, France.,Unité de soutien méthodologique à la recherche clinique, service d'épidémiologie, CHU de Toulouse, Toulouse, France
| | - Adeline Gallini
- INSERM, U1027 and Université Toulouse 3, Toulouse, France.,Unité de soutien méthodologique à la recherche clinique, service d'épidémiologie, CHU de Toulouse, Toulouse, France
| | - Safouane Hamdi
- Laboratoire de Biochimie and Groupe de Recherche en Fertilité Humaine, EA 3694, CHU de Toulouse, Toulouse, France
| | - Pierre Sié
- INSERM, U1048, CHU de Toulouse and Université Toulouse 3, Toulouse Cedex 04, France.,Laboratoire d'Hématologie, CHU de Toulouse, Toulouse Cedex 04, France
| | - Jean-François Arnal
- INSERM, U1048, CHU de Toulouse and Université Toulouse 3, Toulouse Cedex 04, France
| | - Bernard Payrastre
- INSERM, U1048, CHU de Toulouse and Université Toulouse 3, Toulouse Cedex 04, France.,Laboratoire d'Hématologie, CHU de Toulouse, Toulouse Cedex 04, France
| |
Collapse
|
41
|
Valéra MC, Chantalat E, Vinel A, Benoit T, Guillaume M, Game X, Gourdy P, Trémollières F, Payrastre B, Arnal JF. L’association estrogènes et modulateurs sélectifs du récepteur des estrogènes : un renouveau du traitement hormonal ? Presse Med 2015; 44:805-11. [DOI: 10.1016/j.lpm.2015.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/07/2015] [Accepted: 07/08/2015] [Indexed: 11/24/2022] Open
|
42
|
Abot A, Fontaine C, Buscato M, Solinhac R, Flouriot G, Fabre A, Drougard A, Rajan S, Laine M, Milon A, Muller I, Henrion D, Adlanmerini M, Valéra MC, Gompel A, Gerard C, Péqueux C, Mestdagt M, Raymond-Letron I, Knauf C, Ferriere F, Valet P, Gourdy P, Katzenellenbogen BS, Katzenellenbogen JA, Lenfant F, Greene GL, Foidart JM, Arnal JF. The uterine and vascular actions of estetrol delineate a distinctive profile of estrogen receptor α modulation, uncoupling nuclear and membrane activation. EMBO Mol Med 2015; 6:1328-46. [PMID: 25214462 PMCID: PMC4287935 DOI: 10.15252/emmm.201404112] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Estetrol (E4) is a natural estrogen with a long half-life produced only by the human fetal liver during pregnancy. The crystal structures of the estrogen receptor α (ERα) ligand-binding domain bound to 17β-estradiol (E2) and E4 are very similar, as well as their capacity to activate the two activation functions AF-1 and AF-2 and to recruit the coactivator SRC3. In vivo administration of high doses of E4 stimulated uterine gene expression, epithelial proliferation, and prevented atheroma, three recognized nuclear ERα actions. However, E4 failed to promote endothelial NO synthase activation and acceleration of endothelial healing, two processes clearly dependent on membrane-initiated steroid signaling (MISS). Furthermore, E4 antagonized E2 MISS-dependent effects in endothelium but also in MCF-7 breast cancer cell line. This profile of ERα activation by E4, uncoupling nuclear and membrane activation, characterizes E4 as a selective ER modulator which could have medical applications that should now be considered further.
Collapse
Affiliation(s)
- Anne Abot
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Coralie Fontaine
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Mélissa Buscato
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Romain Solinhac
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Gilles Flouriot
- Institut de Recherche en Santé Environnement et Travail, IRSET, INSERM U1085, Team TREC, Biosit, Université de Rennes I, Rennes, France
| | - Aurélie Fabre
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Anne Drougard
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Shyamala Rajan
- Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Muriel Laine
- Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Alain Milon
- CNRS and Université de Toulouse, IPBS, Toulouse, France
| | | | - Daniel Henrion
- INSERM U1083, CNRS UMR 6214, Université d'Angers, Angers, France
| | - Marine Adlanmerini
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Marie-Cécile Valéra
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Anne Gompel
- APHP, Unité de Gynécologie Endocrinienne, Université Paris Descartes, Paris, France
| | - Céline Gerard
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-cancer), Université de Liège, Liège, Belgique
| | - Christel Péqueux
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-cancer), Université de Liège, Liège, Belgique
| | - Mélanie Mestdagt
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-cancer), Université de Liège, Liège, Belgique
| | | | - Claude Knauf
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - François Ferriere
- Institut de Recherche en Santé Environnement et Travail, IRSET, INSERM U1085, Team TREC, Biosit, Université de Rennes I, Rennes, France
| | - Philippe Valet
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Pierre Gourdy
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Benita S Katzenellenbogen
- Departments of Molecular and Integrative Biology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John A Katzenellenbogen
- Departments of Molecular and Integrative Biology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Françoise Lenfant
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Geoffrey L Greene
- Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-cancer), Université de Liège, Liège, Belgique
| | - Jean-François Arnal
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| |
Collapse
|
43
|
Huang J, Shi X, Xi W, Liu P, Long Z, Xi X. Evaluation of targeting c-Src by the RGT-containing peptide as a novel antithrombotic strategy. J Hematol Oncol 2015; 8:62. [PMID: 26025329 PMCID: PMC4459659 DOI: 10.1186/s13045-015-0159-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/22/2015] [Indexed: 01/18/2023] Open
Abstract
Background Interaction of integrin β3 with c-Src plays critical roles in cellular signaling which is heavily implicated in platelet adhesion and aggregation, as well as in tumor cell proliferation and metastasis or in osteoclastic bone resorption. Selectively blocking integrin αIIbβ3 outside-in signaling in platelets has been a focus of attention because of its effective antithrombotic potential together with a sufficient hemostatic capacity. The myristoylated RGT peptide has been shown to achieve this blockade by targeting the association of c-Src with the integrin β3 tail, but the lack of key information regarding the mechanisms of action prevents this strategy from being further developed into practical antithrombotics. Therefore, in-depth knowledge of the precise mechanisms for RGT peptide in regulating platelet function is needed to establish the basis for a potential antithrombotic therapy by targeting c-Src. Methods The reduction-sensitive peptides were applied to rule out the membrane anchorage after cytoplasmic delivery. The c-Src activity was assayed at living cell or at protein levels to assess the direct effect of RGT targeting on c-Src. Thrombus formation under flow in the presence of cytoplasmic RGT peptide was observed by perfusing whole blood through the collagen-coated micro-chamber. Results The RGT peptide did not depend on the membrane anchorage to inhibit outside-in signaling in platelets. The myr-AC ~ CRGT peptide readily blocked agonist-induced c-Src activation by disrupting the Src/β3 association and inhibited the RhoA activation and collagen-induced platelet aggregation in addition to the typical outside-in signaling events. The myr-AC ~ CRGT had no direct effect on the kinase activity of c-Src in living cells as evidenced by its inability to dissociate Csk from c-Src or to alter the phosphorylation level of c-Src Y416 and Y527, consistent results were also from in vitro kinase assays. Under flow conditions, the myr-AC ~ CRGT peptide caused an inhibition of platelet thrombus formation predominantly at high shear rates. Conclusions These findings provide novel insights into the molecular mechanisms by which the RGT peptide regulates integrin signaling and platelet function and reinforce the potential of the RGT peptide-induced disruption of Src/β3 association as a druggable target that would finally enable in vivo and clinical studies using the structure-based small molecular mimetics. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiansong Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| | - Xiaofeng Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| | - Wenda Xi
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| | - Ping Liu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| | - Zhangbiao Long
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| | - Xiaodong Xi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China. .,Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Second Ruijin Road, Shanghai, 200025, China.
| |
Collapse
|
44
|
Burkhart JM, Gambaryan S, Watson SP, Jurk K, Walter U, Sickmann A, Heemskerk JWM, Zahedi RP. What can proteomics tell us about platelets? Circ Res 2014; 114:1204-19. [PMID: 24677239 DOI: 10.1161/circresaha.114.301598] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
More than 130 years ago, it was recognized that platelets are key mediators of hemostasis. Nowadays, it is established that platelets participate in additional physiological processes and contribute to the genesis and progression of cardiovascular diseases. Recent data indicate that the platelet proteome, defined as the complete set of expressed proteins, comprises >5000 proteins and is highly similar between different healthy individuals. Owing to their anucleate nature, platelets have limited protein synthesis. By implication, in patients experiencing platelet disorders, platelet (dys)function is almost completely attributable to alterations in protein expression and dynamic differences in post-translational modifications. Modern platelet proteomics approaches can reveal (1) quantitative changes in the abundance of thousands of proteins, (2) post-translational modifications, (3) protein-protein interactions, and (4) protein localization, while requiring only small blood donations in the range of a few milliliters. Consequently, platelet proteomics will represent an invaluable tool for characterizing the fundamental processes that affect platelet homeostasis and thus determine the roles of platelets in health and disease. In this article we provide a critical overview on the achievements, the current possibilities, and the future perspectives of platelet proteomics to study patients experiencing cardiovascular, inflammatory, and bleeding disorders.
Collapse
Affiliation(s)
- Julia M Burkhart
- From the Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.M.B., A.S., R.P.Z); Institut für Klinische Biochemie und Pathobiochemie, Universitätsklinikum Würzburg, Würzburg, Germany (S.G.); Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia (S.G.); Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.P.W.); Center for Thrombosis and Hemostasis, Universitätsklinikum der Johannes Gutenberg-Universität Mainz, Mainz, Germany (K.J., U.W.); Medizinisches Proteom Center, Ruhr Universität Bochum, Bochum, Germany (A.S.); Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom (A.S.); and Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands (J.W.M.H.)
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: interaction with biological differences between men and women. Br J Pharmacol 2014; 171:580-94. [PMID: 23981051 PMCID: PMC3969074 DOI: 10.1111/bph.12362] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 08/05/2013] [Accepted: 08/16/2013] [Indexed: 12/16/2022] Open
Abstract
Pharmacological response depends on multiple factors and one of them is sex-gender. Data on the specific effects of sex-gender on pharmacokinetics, as well as the safety and efficacy of numerous medications, are beginning to emerge. Nevertheless, the recruitment of women for clinical research is inadequate, especially during the first phases. In general, pharmacokinetic differences between males and females are more numerous and consistent than disparities in pharmacodynamics. However, sex-gender pharmacodynamic differences are now increasingly being identified at the molecular level. It is now even becoming apparent that sex-gender influences pharmacogenomics and pharmacogenetics. Sex-related differences have been reported for several parameters, and it is consistently shown that women have a worse safety profile, with drug adverse reactions being more frequent and severe in women than in men. Overall, the pharmacological status of women is less well studied than that of men and deserves much more attention. The design of clinical and preclinical studies should have a sex-gender-based approach with the aim of tailoring therapies to an individual's needs and concerns.
Collapse
Affiliation(s)
- Flavia Franconi
- Department of Biomedical Sciences, University of SassariSassari, Italy
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
| | - Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
- Department of Surgical, Microsurgical and Medical Sciences, University of SassariSassari, Italy
| |
Collapse
|
46
|
Holmegard HN, Nordestgaard BG, Schnohr P, Tybjaerg-Hansen A, Benn M. Endogenous sex hormones and risk of venous thromboembolism in women and men. J Thromb Haemost 2014; 12:297-305. [PMID: 24329981 DOI: 10.1111/jth.12484] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 12/06/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND Use of oral contraceptives with estrogen and hormone replacement therapy with estrogen or testosterone are associated with increased risk of venous thromboembolism (VTE). However, whether endogenous estradiol and testosterone concentrations are also associated with risk of VTE is unknown. OBJECTIVE We tested the hypothesis that elevated endogenous total estradiol and total testosterone concentrations are associated with increased risk of VTE in the general population. METHODS We studied 4658 women, not receiving exogenous estrogen, and 4673 men from the 1981-1983 Copenhagen City Heart Study, who had estradiol and testosterone concentrations measured. Of these, 636 developed VTE (deep venous thrombosis [DVT] and/or pulmonary embolism [PE]) during a follow-up of 21 years (range, 0.02-32 years). Associations between endogenous estradiol and testosterone concentrations and risk of VTE were estimated by Cox proportional hazards regression with time-dependent covariates and corrected for regression dilution bias. RESULTS Multifactorially adjusted hazard ratios of VTE for individuals with estradiol levels >75th vs. ≤25th percentile were 0.84 (95%CI, 0.25-2.85), 1.05 (0.53-2.08) and 1.05 (0.03-35.13) for pre- and post-menopausal women and men, respectively. For testosterone, corresponding risk estimates were 0.64 (0.03-12.32), 1.11 (0.66-1.86) and 1.30 (0.62-2.73). In addition, no associations were observed between extreme hormone percentiles (>95th vs. ≤75th) and risk of DVT, PE or recurrent VTE. CONCLUSION This prospective study suggests that high endogenous concentrations of estradiol and testosterone in women and men in the general population are not associated with increased risk of VTE, DVT or PE.
Collapse
Affiliation(s)
- H N Holmegard
- Department of Clinical Biochemistry, Gentofte Hospital, Hellerup, Denmark; Copenhagen University Hospitals, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
47
|
Liumbruno GM, Franchini M. Proteomic analysis of venous thromboembolism: an update. Expert Rev Proteomics 2013; 10:179-88. [PMID: 23573784 DOI: 10.1586/epr.13.6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Venous thromboembolism is a complex, multifactorial disorder, the pathogenesis of which typically involves a variety of inherited or acquired factors. The multifactorial etiology of this disease and the partial correlation between genotype and prothrombotic phenotype limit greatly the value of genetic analysis in assessing thrombotic risk. The integration of several new 'omics' techniques enables a multifaceted and holistic approach to the study of venous thrombotic processes and pave the way to the search and identification of novel blood biomarkers and/or effectors of thrombus formation that can also be the possible future target of new anticoagulant and thrombolytic therapies for more personalized medicine. This review provides a comprehensive overview of the latest candidate proteomic biomarkers of venous thrombosis and of the proteomics studies relevant to its pathophysiology, some of which seem to confirm the existence of a common physiopathological basis for venous thromboembolism and atherothrombosis.
Collapse
Affiliation(s)
- Giancarlo Maria Liumbruno
- UOC di Immunoematologia e Medicina Trasfusionale and UOC di Patologia Clinica, San Giovanni Calibita Fatebenefratelli Hospital, 00186 Rome, Italy.
| | | |
Collapse
|
48
|
Arnal JF, Fontaine C, Abot A, Valera MC, Laurell H, Gourdy P, Lenfant F. Lessons from the dissection of the activation functions (AF-1 and AF-2) of the estrogen receptor alpha in vivo. Steroids 2013. [PMID: 23200732 DOI: 10.1016/j.steroids.2012.11.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogens influence most of the physiological processes in mammals, including but not limited to reproduction, cognition, behavior, vascular system, metabolism and bone integrity. Given this widespread role for estrogen in human physiology, it is not surprising that estrogen influence the pathophysiology of numerous diseases, including cancer (of the reproductive tract as breast, endometrial but also colorectal, prostate,…), as well as neurodegenerative, inflammatory-immune, cardiovascular and metabolic diseases, and osteoporosis. These actions are mediated by the activation of estrogen receptors (ER) alpha (ERα) and beta (ERβ), which regulate target gene transcription (genomic action) through two independent activation functions (AF)-1 and AF-2, but can also elicit rapid membrane initiated steroid signals (MISS). Targeted ER gene inactivation has shown that although ERβ plays an important role in the central nervous system and in the heart, ERα appears to play a prominent role in most of the other tissues. Pharmacological activation or inhibition of ERα and/or ERβ provides already the basis for many therapeutic interventions, from hormone replacement at menopause to prevention of the recurrence of breast cancer. However, the use of these estrogens or selective estrogen receptors modulators (SERMs) have also induced undesired effects. Thus, an important challenge consists now to uncouple the beneficial actions from other deleterious ones. The in vivo molecular "dissection" of ERα represents both a molecular and integrated approach that already allowed to delineate in mouse the role of the main "subfunctions" of the receptor and that could pave the way to an optimization of the ER modulation.
Collapse
Affiliation(s)
- Jean-François Arnal
- INSERM U1048-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, 31432 Toulouse, France.
| | | | | | | | | | | | | |
Collapse
|
49
|
|