1
|
Tripathi A, Chhabra A, Rizvi S, Tyagi RK. Selective steroid receptor modulators, degraders and PROTACs: Therapeutic strategies in management of endocrine-related cancers. Mol Cell Endocrinol 2025; 605:112569. [PMID: 40339978 DOI: 10.1016/j.mce.2025.112569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/23/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Endocrine-related disorders are highly prevalent globally, affecting millions of people. Such diseases are multifactorial in origin and are influenced by the complex interplay of genetics, lifestyle, and environmental factors. Recurring disruptions in the endocrine homeostasis can lead to a cascade of endocrine-related cancers. It is well known that nuclear receptors (NRs), particularly estrogen receptor and androgen receptor malfunctioning promote the oncogenesis of breast cancer and prostate cancer, respectively. However, existing therapeutics against these diseases, including aromatase inhibitors, (anti-) hormonal therapy, etc., often yield limited success, prompting to explore alternative methods of disease management. Additionally, drug resistance is prominent in cancer patients undergoing multidrug therapy. Currently, novel drug design strategies targeting NRs are being implemented for the discovery of a new generation of small molecule modulators, including selective NR modulators (SNuRMs) and degraders (SNuRDs). Moreover, proteolysis-targeting chimeras (PROTACs) as NR degraders, are also being developed primarily to overcome drug resistance, enhance protein selectivity, and mitigate off-target toxicity. This review highlights recent advancements in SNuRMs and SNuRDs for managing NRs-associated endocrine/metabolic disorders. Furthermore, we discuss the therapeutic potential of PROTAC degraders as a stand-alone strategy for receptor-mediated disease intervention, offering new avenues for precision medicine.
Collapse
Affiliation(s)
- Anjali Tripathi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Ayushi Chhabra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Sheeba Rizvi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India.
| |
Collapse
|
2
|
Xia W, Khalil RA. Hormone Replacement Therapy and Cardiovascular Health in Postmenopausal Women. Int J Mol Sci 2025; 26:5078. [PMID: 40507889 PMCID: PMC12154064 DOI: 10.3390/ijms26115078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/16/2025] [Accepted: 05/10/2025] [Indexed: 06/16/2025] Open
Abstract
Sex-related differences are found not only in the reproductive system but also across various biological systems, such as the cardiovascular system. Compared with premenopausal women, cardiovascular disease (CVD) tends to occur more frequently in adult men and postmenopausal women (Post-MW). Also, during the reproductive years, sex hormones synthesized and released into the blood stream affect vascular function in a sex-dependent fashion. Estrogen (E2) interacts with estrogen receptors (ERs) in endothelial cells, vascular smooth muscle, and the extracellular matrix, causing both genomic and non-genomic effects, including vasodilation, decreased blood pressure, and cardiovascular protection. These observations have suggested beneficial effects of female sex hormones on cardiovascular function. In addition, the clear advantages of E2 supplementation in alleviating vasomotor symptoms during menopause have led to clinical investigations of the effects of menopausal hormone therapy (MHT) in CVD. However, the findings from these clinical trials have been variable and often contradictory. The lack of benefits of MHT in CVD has been related to the MHT preparation (type, dose, and route), vascular ERs (number, variants, distribution, and sensitivity), menopausal stage (MHT timing, initiation, and duration), hormonal environment (progesterone, testosterone (T), gonadotropins, and sex hormone binding globulin), and preexisting cardiovascular health and other disorders. The vascular effects of sex hormones have also prompted further examination of the use of anabolic drugs among athletes and the long-term effects of E2 and T supplements on cardiovascular health in cis- and transgender individuals seeking gender-affirming therapy. Further analysis of the effects of sex hormones and their receptors on vascular function should enhance our understanding of the sex differences and menopause-related changes in vascular signaling and provide better guidance for the management of CVD in a gender-specific fashion and in Post-MW.
Collapse
Affiliation(s)
| | - Raouf A. Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
3
|
Costa TJ, Fontes MT, Barros PR, Hope MC, Webb RC, Wenceslau CF, Enos RT, McCarthy CG. Overexpression of adipose tissue ERα enhances PVAT anticontractility via NOX4-derived H 2O 2 and is protective against high-fat diet-induced dysfunction. Am J Physiol Heart Circ Physiol 2025; 328:H1065-H1072. [PMID: 40127093 DOI: 10.1152/ajpheart.00180.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Menopause has unequivocally been associated with cardiovascular risk and obesity. Loss of estrogen bioavailability is a hallmark of menopause. Estrogen is generally considered vasculoprotective, with estrogen receptor α (ERα) being the predominant receptor subtype that mediates these positive effects. Similarly, estrogen and ERα are known to stimulate white adipose tissue metabolism. However, it is unknown whether ERα could exert a beneficial effect on mesenteric perivascular adipose tissue (PVAT). PVAT is a heterogeneous tissue that surrounds most peripheral blood vessels. In physiological conditions, PVAT has an anticontractile effect on the vasculature. However, in several diseases, PVAT switches its phenotype to become procontractile. To date, the role of ERα in PVAT function in health and disease is unknown. Therefore, we hypothesized that overexpression of adipose tissue ERα (ERαOE) would 1) increase the anticontractile effect of PVAT in chow diet conditions and 2) protect mice against a high-fat diet (HFD)-induced PVAT dysfunction. To test this hypothesis, mesenteric resistance arteries, with and without PVAT, were isolated from female ERαOE mice, which had either been on a regular chow diet or an HFD for 19 wk. We observed that ERαOE amplifies the anticontractile effect of mesenteric PVAT via NADPH oxidase 4 (NOX4)-derived hydrogen peroxide (H2O2) in chow conditions, and ERαOE is protective against a dysfunctional PVAT that is observed after an HFD, via the same anticontractile mechanism. Collectively, these data demonstrate that ERα is vasculoprotective in the context of PVAT. Harnessing this signaling could be important for reducing cardiovascular risk in postmenopausal women.NEW & NOTEWORTHY We have revealed for the first time that overexpression of adipose tissue estrogen receptor α (ERαOE) amplifies the anticontractile effect of mesenteric PVAT via the biosynthesis of NADPH oxidase 4 (NOX4)-derived hydrogen peroxide (H2O2), and this overexpression is protective against HFD-induced PVAT dysfunction. Collectively, these data demonstrate an important mechanism by which ERα signaling is vasculoprotective in the context of PVAT.
Collapse
Affiliation(s)
- Tiago J Costa
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Milene T Fontes
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Paula R Barros
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Marion C Hope
- Department of Pathology, Microbiology, & Immunology, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, United States
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Reilly T Enos
- Department of Pathology, Microbiology, & Immunology, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| |
Collapse
|
4
|
Li T, Thoen ZE, Applebaum JM, Khalil RA. Menopause-related changes in vascular signaling by sex hormones. J Pharmacol Exp Ther 2025; 392:103526. [PMID: 40184819 DOI: 10.1016/j.jpet.2025.103526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/07/2025] Open
Abstract
Cardiovascular disease (CVD), such as hypertension and coronary artery disease, involves pathological changes in vascular signaling, function, and structure. Vascular signaling is regulated by multiple intrinsic and extrinsic factors that influence endothelial cells, vascular smooth muscle, and extracellular matrix. Vascular function is also influenced by environmental factors including diet, exercise, and stress, as well as genetic background, sex differences, and age. CVD is more common in adult men and postmenopausal women than in premenopausal women. Specifically, women during menopausal transition, with declining ovarian function and production of estrogen (E2) and progesterone, show marked increase in the incidence of CVD and associated vascular dysfunction. Mechanistic research suggests that E2 and E2 receptor signaling have beneficial effects on vascular function including vasodilation, decreased blood pressure, and cardiovascular protection. Also, the tangible benefits of E2 supplementation in improving menopausal symptoms have prompted clinical trials of menopausal hormone therapy (MHT) in CVD, but the results have been inconsistent. The inadequate benefits of MHT in CVD could be attributed to the E2 type, dose, formulation, route, timing, and duration as well as menopausal changes in E2/E2 receptor vascular signaling. Other factors that could affect the responsiveness to MHT are the integrated hormonal milieu including gonadotropins, progesterone, and testosterone, vascular health status, preexisting cardiovascular conditions, and menopause-related dysfunction in the renal, gastrointestinal, endocrine, immune, and nervous systems. Further analysis of these factors should enhance our understanding of menopause-related changes in vascular signaling by sex hormones and provide better guidance for management of CVD in postmenopausal women. SIGNIFICANCE STATEMENT: Cardiovascular disease is more common in adult men and postmenopausal women than premenopausal women. Earlier observations of vascular benefits of menopausal hormone therapy did not materialize in randomized clinical trials. Further examination of the cardiovascular effects of sex hormones in different formulations and regimens, and the menopausal changes in vascular signaling would help to adjust the menopausal hormone therapy protocols in order to enhance their effectiveness in reducing the risk and the management of cardiovascular disease in postmenopausal women.
Collapse
Affiliation(s)
- Tao Li
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Zachary E Thoen
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jessica M Applebaum
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
5
|
Nicolas J, Nicolas J, Nicolas A, Safieddine M, Gannage Yared MH. Retrospective and longitudinal study of blood pressure in school-aged children. Clin Nutr ESPEN 2025; 66:76-82. [PMID: 39824456 DOI: 10.1016/j.clnesp.2025.01.030] [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: 11/19/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND Pediatric hypertension (HTN) is increasing over time. Worldwide, the blood pressure (BP) trend has been poorly studied. The aim of our study is to evaluate the recent trend in BP in a school-aged children sample, and to search for factors that might influence BP variability. MATERIALS AND METHODS This is a retrospective longitudinal analysis studying the BP of school-aged children covering the classes of 2013-2021. From medical records of the children, sex, date of birth, date of medical visit which takes place every two years (in the 6th, 8th, and 10th grade), height (in cm), weight (in kg), systolic blood pressure (SBP), and diastolic blood pressure (DBP), were collected for each child. RESULTS The sample consisted of 2164 children. SBP and DBP were significantly lower in girls than in boys and were significantly correlated with body mass index (BMI) (p-value <0.001 for all comparisons). A significant increase in SBP and DBP was observed from the 6th to the 10th grade, as well as a significant decrease of BP during a nine-year follow-up (p-values <0.001 for all comparisons). CONCLUSION Our results confirmed a sex difference in BP, an increase in BP with age and a positive association between BMI and BP. Finally, a negative secular trend for BP was found. Further studies are needed to determine if this decrease in BP with time will persist after the coronavirus disease 19 pandemic.
Collapse
Affiliation(s)
- Jad Nicolas
- Hotel-Dieu de France, Department of Endocrinology, Saint-Joseph University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Jessica Nicolas
- Hotel-Dieu de France, Department of Endocrinology, Saint-Joseph University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Anthony Nicolas
- Hotel-Dieu de France, Department of Endocrinology, Saint-Joseph University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Maissa Safieddine
- Hotel-Dieu de France, Clinical Research Center, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Marie-Helene Gannage Yared
- Hotel-Dieu de France, Department of Endocrinology, Saint-Joseph University of Beirut, Faculty of Medicine, Beirut, Lebanon.
| |
Collapse
|
6
|
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] [Download PDF] [Figures] [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
|
7
|
Amin YA, Elnegiry AA, Awadalla EA, Hussein HA, Mohamed RH. Placental expression of estrogen and progesterone receptors and vascular endothelial growth factor in buffaloes suffering from uterine torsion. BMC Vet Res 2025; 21:65. [PMID: 39953531 PMCID: PMC11827448 DOI: 10.1186/s12917-024-04422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/02/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Although several risk factors have been suggested for uterine torsion, the pathogenesis is still unclear. Therefore, the current study aims to investigate the pathogenesis of uterine torsion by assessing the histological, histochemical, and immunohistochemical changes that occur in the placenta obtained from uterine torsion cases. Immunohistochemical changes include investigation of the expression of estrogen receptors (ERs), progesterone receptors (PRs), and the vascular endothelial growth factor (VEGF) in the placental tissue. METHODS Forty intrapartum dairy cows were included in this investigation. The cows were divided into two equal groups. The first group was the uterine torsion (UT) group, while the second group was the normal control group (Ctrl). After caesarian section treatment, placentas were collected from all animals in the study. Histopathological, histochemical, and immunohistochemical examinations were performed. Estrogen receptors, progesterone receptors, and vascular endothelial growth factor expression in the placenta were evaluated. RESULTS The results revealed numerous trophoblast giant or binucleate cells in the trophoblastic epithelium. Through Masson's trichrome technique, the distribution of collagen fibers as shiny, blue-colored stripes on the fetal mesenchyme was observed. Additionally, the results showed a strong, intense PAS-positive reaction in the cytoplasmic vesicles of most trophoblastic cells due to mucopolysaccharides. The immunohistochemical findings of the UT placenta revealed moderate to weak staining for ERs in contrast to those of the Ctrl placenta, which revealed moderate staining for ERs. In addition, non-statistical differences in the expression of PRs were found between the two tested groups. For VEGF, strong positive immunoreactivity was found in the Ctrl group compared to the UT group, which exhibits a general absence in many trophoblast cells. CONCLUSION It can be concluded that significant variation was observed in the placentas obtained from buffaloes suffering from UT compared to those obtained from normal pregnant ones. These significant variations were involved in the decreased expression of ERs and VEGF in the UT group compared to the normal Ctrl one. Investigating the expression of these placental molecules may monitor the changes in the placental tissue and provide insight into the pathogenesis of UT.
Collapse
Affiliation(s)
- Yahia A Amin
- Department of Theriogenology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81528, Egypt.
| | - Ahmed Abdou Elnegiry
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Eatemad A Awadalla
- Department of Zoology, Faculty of Science, Aswan University, Aswan, 81528, Egypt
| | - Hassan A Hussein
- Department of Theriogenology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
- Faculty of Veterinary Medicine, Sphinx University, New Assiut, Egypt
| | - Ragab H Mohamed
- Department of Theriogenology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81528, Egypt
| |
Collapse
|
8
|
Gonzalez G, Lin X, Herath I, Veselkov K, Bronstein M, Zitnik M. Combinatorial prediction of therapeutic perturbations using causally-inspired neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.03.573985. [PMID: 38260532 PMCID: PMC10802439 DOI: 10.1101/2024.01.03.573985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Phenotype-driven approaches identify disease-counteracting compounds by analyzing the phenotypic signatures that distinguish diseased from healthy states. These approaches can guide the discovery of targeted perturbations, including small-molecule drugs and genetic interventions, that modulate disease phenotypes toward healthier states. Here, we introduce PDGrapher, a causally inspired graph neural network (GNN) designed to predict combinatorial perturbagens (sets of therapeutic targets) capable of reversing disease phenotypes. Unlike methods that learn how perturbations alter phenotypes, PDGrapher solves the inverse problem of directly predicting the perturbagens needed to achieve a desired response. PDGrapher is a GNN that embeds disease cell states into gene regulatory or protein-protein interaction networks, learns a latent representation of these states, and identifies the optimal combinatorial perturbations that most effectively shift the diseased state toward the desired treated state within that latent space. In experiments in nine cell lines with chemical perturbations, PDGrapher identified effective per-turbagens in up to 13.33% more test samples than competing methods and achieved a normalized discounted cumulative gain of up to 0.12 higher to classify therapeutic targets. It also demonstrated competitive performance on ten genetic perturbation datasets. A key advantage of PDGrapher is its direct prediction paradigm, in contrast to the indirect and computationally intensive models traditionally employed in phenotype-driven research. This approach accelerates training by up to 25 times compared to existing methods. PDGrapher provides a fast approach for identifying therapeutic perturbations and advancing phenotype-driven drug discovery.
Collapse
Affiliation(s)
- Guadalupe Gonzalez
- Imperial College London, London, UK
- Prescient Design, Genentech, South San Francisco, CA, USA
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xiang Lin
- Harvard Medical School, Boston, MA, USA
| | - Isuru Herath
- Merck & Co., South San Francisco, CA, USA
- Cornell University, Ithaca, NY, USA
| | | | | | - Marinka Zitnik
- Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
9
|
Tondo P, Meschi C, Mantero M, Scioscia G, Siciliano M, Bradicich M, Stella GM. Sex and gender differences during the lung lifespan: unveiling a pivotal impact. Eur Respir Rev 2025; 34:240121. [PMID: 39971394 PMCID: PMC11836673 DOI: 10.1183/16000617.0121-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/29/2024] [Indexed: 02/21/2025] Open
Abstract
Sex and gender differences significantly influence lung parenchyma development, beginning as early as the embryonic stages of human life. Although this association is well known in the clinical manifestations of some relevant pulmonary diseases, there is less data available regarding their effects on cell biological programmes across different stages of body development. A deep understanding of these mechanisms could help in defining preventive strategies tailored to a fully personalised approach to respiratory medicine. From this perspective, this review aims to analyse the influence of sex and gender on bronchoalveolar and vascular compartments from embryonic and neonatal stages through to adolescence, adulthood and elder age.
Collapse
Affiliation(s)
- Pasquale Tondo
- Department of Medical and Surgical Sciences, University of Foggia; Respiratory and Critical Care Unit, Polyclinic Foggia University-Hospital, Foggia, Italy
| | - Claudia Meschi
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
- Pulmonary Unit, Cardiothoracic and Vascular Department, Pisa University Hospital, Pisa, Italy
| | - Marco Mantero
- Department of Health Sciences, Respiratory Unit, ASST Santi Paolo e Carlo, San Paolo Hospital, University of Milan, Milan, Italy
| | - Giulia Scioscia
- Department of Medical and Surgical Sciences, University of Foggia; Respiratory and Critical Care Unit, Polyclinic Foggia University-Hospital, Foggia, Italy
| | - Matteo Siciliano
- Catholic University of the Sacred Heart, Rome Campus; Agostino Gemelli IRCCS University Polyclinic Foundation, Rome, Italy
| | - Matteo Bradicich
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Giulia M. Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS San Matteo Polyclinic Hospital, Pavia, Italy
| |
Collapse
|
10
|
Bhullar SK, Rabinovich-Nikitin I, Kirshenbaum LA. Oral hormonal contraceptives and cardiovascular risks in females. Can J Physiol Pharmacol 2024; 102:572-584. [PMID: 38781602 DOI: 10.1139/cjpp-2024-0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Oral hormonal contraception (OHC) is a widely employed method in females for the prevention of unintended pregnancies, as well as for the treatment of menstrual disorders, endometriosis, and polycystic ovarian syndrome. However, it is believed that with OHCs use, some females may have higher risk of cardiovascular diseases, such as hypertension, diabetes, myocardial infarction, thrombosis, and heart failure. Although such risks are infrequently detected in healthy young females with the use of oral contraceptives, slightly elevated risks of cardiovascular diseases have been observed among reproductive-aged healthy females. However, prolonged use of OHC has also been claimed to have protective cardiac effects and may contribute to reduced risk of cardiovascular disease. In fact, the debate on whether OHC administration increases the risk of cardiovascular diseases has been ongoing with inconsistent and controversial viewpoints. Nevertheless, a great deal of work has been carried out to understand the relationship between OHC use and the occurrence of cardiovascular risk in females who use OHC for preventing the unwanted pregnancy or treatment of other disorders. Therefore, in this review we summarize the most recent available evidence regarding the association between the use of oral hormonal contraceptives and the risk for cardiovascular disease in females who are using OHC to prevent unintended pregnancy.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Inna Rabinovich-Nikitin
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lorrie A Kirshenbaum
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
11
|
Delgado Spicuzza JM, Gosalia J, Zhong L, Bondonno C, Petersen KS, De Souza MJ, Alipour E, Kim-Shapiro DB, Somani YB, Proctor DN. Seven-day dietary nitrate supplementation clinically significantly improves basal macrovascular function in postmenopausal women: a randomized, placebo-controlled, double-blind, crossover clinical trial. Front Nutr 2024; 11:1359671. [PMID: 38915856 PMCID: PMC11194363 DOI: 10.3389/fnut.2024.1359671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/09/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction Cardiovascular disease (CVD) is the leading cause of death in women, with increased risk following menopause. Dietary intake of beetroot juice and other plant-based nitrate-rich foods is a promising non-pharmacological strategy for increasing systemic nitric oxide and improving endothelial function in elderly populations. The purpose of this randomized, placebo-controlled, double-blind, crossover clinical trial was to determine the effects of short-term dietary nitrate (NO3 -) supplementation, in the form of beetroot juice, on resting macrovascular endothelial function and endothelial resistance to whole-arm ischemia-reperfusion (IR) injury in postmenopausal women at two distinct stages of menopause. Methods Early-postmenopausal [1-6 years following their final menstrual period (FMP), n = 12] and late-postmenopausal (6+ years FMP, n = 12) women consumed nitrate-rich (400 mg NO3 -/70 mL) and nitrate-depleted beetroot juice (approximately 40 mg NO3 -/70 mL, placebo) daily for 7 days. Brachial artery flow-mediated dilation (FMD) was measured pre-supplementation (Day 0), and approximately 24 h after the last beetroot juice (BR) dose (Day 8, post-7-day BR). Consequently, FMD was measured immediately post-IR injury and 15 min later (recovery). Results Results of the linear mixed-effects model revealed a significantly greater increase in resting FMD with 7 days of BRnitrate compared to BRplacebo (mean difference of 2.21, 95% CI [0.082, 4.34], p = 0.042); however, neither treatment blunted the decline in post-IR injury FMD in either postmenopausal group. Our results suggest that 7-day BRnitrate-mediated endothelial protection is lost within the 24-h period following the final dose of BRnitrate. Conclusion Our findings demonstrate that nitrate-mediated postmenopausal endothelial protection is dependent on the timing of supplementation in relation to IR injury and chronobiological variations in dietary nitrate metabolism. Clinical trial registration https://classic.clinicaltrials.gov/ct2/show/NCT03644472.
Collapse
Affiliation(s)
- Jocelyn M. Delgado Spicuzza
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Jigar Gosalia
- Integrative Vascular Physiology Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Liezhou Zhong
- Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Catherine Bondonno
- Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Kristina S. Petersen
- Cardiometabolic Nutrition Research Lab, Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Mary Jane De Souza
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
- Women’s Health and Exercise Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, NC, United States
| | | | - Yasina B. Somani
- Faculty of Biological Sciences, Department of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - David N. Proctor
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
- Integrative Vascular Physiology Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
12
|
Li C, Ajmal E, Alok K, Powell K, Wadolowski S, Tambo W, Turpin J, Barthélemy E, Al-Abed Y, LeDoux D. CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury. Biol Sex Differ 2024; 15:44. [PMID: 38816868 PMCID: PMC11138127 DOI: 10.1186/s13293-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression. METHODS Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes. RESULTS Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males. CONCLUSIONS This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.
Collapse
Affiliation(s)
- Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA.
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Khaled Alok
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Steven Wadolowski
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Justin Turpin
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Ernest Barthélemy
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - David LeDoux
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| |
Collapse
|
13
|
Weiner CM, Khan SE, Leong C, Ranadive SM, Campbell SC, Howard JT, Heffernan KS. Association of enterolactone with blood pressure and hypertension risk in NHANES. PLoS One 2024; 19:e0302254. [PMID: 38743749 PMCID: PMC11093351 DOI: 10.1371/journal.pone.0302254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/30/2024] [Indexed: 05/16/2024] Open
Abstract
The gut microbiome may affect overall cardiometabolic health. Enterolactone is an enterolignan reflective of dietary lignan intake and gut microbiota composition and diversity that can be measured in the urine. The purpose of this study was to examine the association between urinary enterolactone concentration as a reflection of gut health and blood pressure/risk of hypertension in a large representative sample from the US population. This analysis was conducted using data from the National Health and Nutrition Examination Survey (NHANES) collected from January 1999 through December 2010. Variables of interest included participant characteristics (including demographic, anthropometric and social/environmental factors), resting blood pressure and hypertension history, and urinary enterolactone concentration. 10,637 participants (45 years (SE = 0.3), 51.7% (SE = 0.6%) were female) were included in analyses. In multivariable models adjusted for demographic, socioeconomic and behavioral/environmental covariates, each one-unit change in log-transformed increase in enterolactone was associated with a 0.738 point (95% CI: -0.946, -0.529; p<0.001) decrease in systolic blood pressure and a 0.407 point (95% CI: -0.575, -0.239; p<0.001) decrease in diastolic blood pressure. Moreover, in fully adjusted models, each one-unit change in log-transformed enterolactone was associated with 8.2% lower odds of hypertension (OR = 0.918; 95% CI: 0.892, 0.944; p<0.001). Urinary enterolactone, an indicator of gut microbiome health, is inversely associated with blood pressure and hypertension risk in a nationally representative sample of U.S. adults.
Collapse
Affiliation(s)
- Cynthia M. Weiner
- Department of Kinesiology, University of Maryland, College Park, Maryland, United States of America
| | - Shannon E. Khan
- Department of Kinesiology, University of Maryland, College Park, Maryland, United States of America
| | - Caleb Leong
- Department of Public Health, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas, United States of America
| | - Sushant M. Ranadive
- Department of Kinesiology, University of Maryland, College Park, Maryland, United States of America
| | - Sara C. Campbell
- Department of Kinesiology and Health, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Jeffrey T. Howard
- Department of Public Health, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas, United States of America
| | - Kevin S. Heffernan
- Department of Exercise Science, Syracuse University, Syracuse, NY, United States of America
| |
Collapse
|
14
|
Vidal-dos-Santos M, Anunciação LF, Armstrong-Jr R, Ricardo-da-Silva FY, Ramos IYT, Correia CJ, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. 17β-estradiol and methylprednisolone association as a therapeutic option to modulate lung inflammation in brain-dead female rats. Front Immunol 2024; 15:1375943. [PMID: 38765005 PMCID: PMC11099279 DOI: 10.3389/fimmu.2024.1375943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/15/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction Brain death (BD) is known to compromise graft quality by causing hemodynamic, metabolic, and hormonal changes. The abrupt reduction of female sex hormones after BD was associated with increased lung inflammation. The use of both corticoids and estradiol independently has presented positive results in modulating BD-induced inflammatory response. However, studies have shown that for females the presence of both estrogen and corticoids is necessary to ensure adequate immune response. In that sense, this study aims to investigate how the association of methylprednisolone (MP) and estradiol (E2) could modulate the lung inflammation triggered by BD in female rats. Methods Female Wistar rats (8 weeks) were divided into four groups: sham (animals submitted to the surgical process, without induction of BD), BD (animals submitted to BD), MP/E2 (animals submitted to BD that received MP and E2 treatment 3h after BD induction) and MP (animals submitted to BD that received MP treatment 3h after BD induction). Results Hemodynamics, systemic and local quantification of IL-6, IL-1β, VEGF, and TNF-α, leukocyte infiltration to the lung parenchyma and airways, and adhesion molecule expression were analyzed. After treatment, MP/E2 association was able to reinstate mean arterial pressure to levels close to Sham animals (p<0.05). BD increased leukocyte infiltration to the airways and MP/E2 was able to reduce the number of cells (p=0.0139). Also, the associated treatment modulated the vasculature by reducing the expression of VEGF (p=0.0616) and maintaining eNOS levels (p=0.004) in lung tissue. Discussion Data presented in this study show that the association between corticoids and estradiol could represent a better treatment strategy for lung inflammation in the female BD donor by presenting a positive effect in the hemodynamic management of the donor, as well as by reducing infiltrated leukocyte to the airways and release of inflammatory markers in the short and long term.
Collapse
Affiliation(s)
- Marina Vidal-dos-Santos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Lucas F. Anunciação
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Armstrong-Jr
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Y. Ricardo-da-Silva
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isabella Yumi Taira Ramos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano J. Correia
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz F. P. Moreira
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Henri G. D. Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Ana C. Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Hu C, Lu L, Guo C, Zhan T, Zhang X, Zhang H. Bisphenols and brominated bisphenols induced endothelial dysfunction via its disruption of endothelial nitric oxide synthase. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123600. [PMID: 38369087 DOI: 10.1016/j.envpol.2024.123600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/20/2024]
Abstract
Emerging literatures have concentrated on the association between cardiovascular diseases risk of typical endocrine disruptor bisphenols, which also put forward the further studies need respect to the potential mechanism. Herein, we investigated the endothelial dysfunction effects of bisphenols and brominated bisphenols involved in aortic pathological structure, endothelial nitric oxide synthase (eNOS) protein phosphorylation, synthase activity and nitric oxide (NO) production in human umbilical vein endothelial cells (HUVECs) and C57BL/6 mice. Bisphenol A (BPA) and bisphenol S (BPS) increased NO production by 85.7% and 68.8% at 10-6 M level in vitro and 74.3%, 41.5% in vivo, respectively, while tetrabromobisphenol S (TBBPS) significantly inhibited NO by 55.7% at 10-6 M in vitro and 28.9% in vivo at dose of 20 mg/kg BW/d. Aortic transcriptome profiling revealed that the process of 'regulation of NO mediated signal transduction' was commonly induced. The mRNA and protein expression of phosphorylated eNOS at Ser1177 were promoted by BPA and BPS but decreased by TBBPA and TBBPS in HUVECs. Phosphorylation and enzymatic activity of eNOS were significantly increased by 43.4% and 13.8% with the treatment of BPA and BPS at 10-7 M, but decreased by 16.9% after exposure to TBBPS at 10-6 M in vitro. Moreover, only TBBPS was observed to increase aorta thickness significantly in mice and induce endothelial dysfunction. Our work suggests that bisphenols and brominated bisphenols may exert adverse outcome on vascular health differently in vitro and in vivo, and emphasizes areas of public health concern similar endocrine disruptors vulnerable on the vascular endothelial function.
Collapse
Affiliation(s)
- Chao Hu
- Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Liping Lu
- Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Hangzhou International Urbanology Research Center and Center for Zhejiang Urban Governance Studies, Hangzhou, 311121, China.
| | - Chunyan Guo
- Radiation Monitoring Technical Center, State Environmental Protection Key Laboratory of Radiation Environmental Monitoring, Ministry of Ecology and Environment, Hangzhou, 310012, China
| | - Tingjie Zhan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ, 08854, United States
| | - Xiaofang Zhang
- Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Hangjun Zhang
- Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Hangzhou International Urbanology Research Center and Center for Zhejiang Urban Governance Studies, Hangzhou, 311121, China
| |
Collapse
|
16
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
17
|
Collignon A, Dion-Albert L, Ménard C, Coelho-Santos V. Sex, hormones and cerebrovascular function: from development to disorder. Fluids Barriers CNS 2024; 21:2. [PMID: 38178239 PMCID: PMC10768274 DOI: 10.1186/s12987-023-00496-3] [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: 08/11/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
Proper cerebrovascular development and neurogliovascular unit assembly are essential for brain growth and function throughout life, ensuring the continuous supply of nutrients and oxygen. This involves crucial events during pre- and postnatal stages through key pathways, including vascular endothelial growth factor (VEGF) and Wnt signaling. These pathways are pivotal for brain vascular growth, expansion, and blood-brain barrier (BBB) maturation. Interestingly, during fetal and neonatal life, cerebrovascular formation coincides with the early peak activity of the hypothalamic-pituitary-gonadal axis, supporting the idea of sex hormonal influence on cerebrovascular development and barriergenesis.Sex hormonal dysregulation in early development has been implicated in neurodevelopmental disorders with highly sexually dimorphic features, such as autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD). Both disorders show higher prevalence in men, with varying symptoms between sexes, with boys exhibiting more externalizing behaviors, such as aggressivity or hyperactivity, and girls displaying higher internalizing behaviors, including anxiety, depression, or attention disorders. Indeed, ASD and ADHD are linked to high prenatal testosterone exposure and reduced aromatase expression, potentially explaining sex differences in prevalence and symptomatology. In line with this, high estrogen levels seem to attenuate ADHD symptoms. At the cerebrovascular level, sex- and region-specific variations of cerebral blood flow perfusion have been reported in both conditions, indicating an impact of gonadal hormones on the brain vascular system, disrupting its ability to respond to neuronal demands.This review aims to provide an overview of the existing knowledge concerning the impact of sex hormones on cerebrovascular formation and maturation, as well as the onset of neurodevelopmental disorders. Here, we explore the concept of gonadal hormone interactions with brain vascular and BBB development to function, with a particular focus on the modulation of VEGF and Wnt signaling. We outline how these pathways may be involved in the underpinnings of ASD and ADHD. Outstanding questions and potential avenues for future research are highlighted, as uncovering sex-specific physiological and pathological aspects of brain vascular development might lead to innovative therapeutic approaches in the context of ASD, ADHD and beyond.
Collapse
Affiliation(s)
- Adeline Collignon
- Department of Psychiatry & Neuroscience and CERVO Brain Research Center, Universite Laval, Quebec City, Canada
| | - Laurence Dion-Albert
- Department of Psychiatry & Neuroscience and CERVO Brain Research Center, Universite Laval, Quebec City, Canada
| | - Caroline Ménard
- Department of Psychiatry & Neuroscience and CERVO Brain Research Center, Universite Laval, Quebec City, Canada
| | - Vanessa Coelho-Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, Institute of Physiology, Coimbra, Portugal.
| |
Collapse
|
18
|
Ashraf H, Ashfaq H, Ashraf A. Gender and racial disparities in obesity-related cardiovascular-induced mortality in the USA, 1999-2020. Curr Probl Cardiol 2024; 49:102178. [PMID: 37907185 DOI: 10.1016/j.cpcardiol.2023.102178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Obesity is closely linked to various cardiovascular diseases, leading to increased mortality rates. This study examines the trends in cardiovascular-induced obesity associated mortality among individuals aged 15 years and older in the United States. Data were sourced from the CDC WONDER for the years 1999-2020, encompassing fatalities where cardiovascular disease was the underlying cause of death and obesity was a contributing factor. The dataset was analyzed, considering annual patterns, gender, and ethnic origins. A total of 280,992 deaths were reported, with 56.35 % attributed to males and 43.64 % to females. The age-adjusted mortality rate was 5.8 for males and 4 for females. Non-Hispanic white individuals accounted for 71.049 % of total deaths, while non-Hispanic Black individuals contributed 19.510 %. The highest mortality rate was observed among non-Hispanic Black individuals, with non-Hispanic American Indian or Alaska Native individuals following. Non-Hispanic Asian or Pacific Islander individuals had the lowest mortality rate. The average annual percentage change (AAPC) was 6.1 for males and 4.4 for females. A significant increase in the overall mortality rate was observed from 2018 to 2020, with Hispanics/Latinos exhibiting the highest increase. The elevated AAMR among males as compared to females may be attributed to the cardio-protective properties of estrogen in women. Furthermore, the COVID-19 pandemic introduced unfavorable lifestyle changes, including weight gain and reduced exercise, potentially exacerbating CVD mortality trends after 2019. Further timely and targeted efforts are needed to control obesity and cardiovascular-related mortality in the USA.
Collapse
Affiliation(s)
- Hamza Ashraf
- Department of Cardiology, Allama Iqbal Medical College, Lahore, Pakistan.
| | - Haider Ashfaq
- Department of Medicine, Allama Iqbal Medical College, Lahore, Pakistan
| | - Ali Ashraf
- Government College University, Lahore, Pakistan
| |
Collapse
|
19
|
Drury ER, Wu J, Gigliotti JC, Le TH. Sex differences in blood pressure regulation and hypertension: renal, hemodynamic, and hormonal mechanisms. Physiol Rev 2024; 104:199-251. [PMID: 37477622 PMCID: PMC11281816 DOI: 10.1152/physrev.00041.2022] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/06/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023] Open
Abstract
The teleology of sex differences has been argued since at least as early as Aristotle's controversial Generation of Animals more than 300 years BC, which reflects the sex bias of the time to contemporary readers. Although the question "why are the sexes different" remains a topic of debate in the present day in metaphysics, the recent emphasis on sex comparison in research studies has led to the question "how are the sexes different" being addressed in health science through numerous observational studies in both health and disease susceptibility, including blood pressure regulation and hypertension. These efforts have resulted in better understanding of differences in males and females at the molecular level that partially explain their differences in vascular function and renal sodium handling and hence blood pressure and the consequential cardiovascular and kidney disease risks in hypertension. This review focuses on clinical studies comparing differences between men and women in blood pressure over the life span and response to dietary sodium and highlights experimental models investigating sexual dimorphism in the renin-angiotensin-aldosterone, vascular, sympathetic nervous, and immune systems, endothelin, the major renal sodium transporters/exchangers/channels, and the impact of sex hormones on these systems in blood pressure homeostasis. Understanding the mechanisms governing sex differences in blood pressure regulation could guide novel therapeutic approaches in a sex-specific manner to lower cardiovascular risks in hypertension and advance personalized medicine.
Collapse
Affiliation(s)
- Erika R Drury
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Jing Wu
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, United States
| | - Joseph C Gigliotti
- Department of Integrative Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia, United States
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
20
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [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: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
21
|
Youngblood H, Schoenlein PV, Pasquale LR, Stamer WD, Liu Y. Estrogen dysregulation, intraocular pressure, and glaucoma risk. Exp Eye Res 2023; 237:109725. [PMID: 37956940 PMCID: PMC10842791 DOI: 10.1016/j.exer.2023.109725] [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: 09/01/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Characterized by optic nerve atrophy due to retinal ganglion cell (RGC) death, glaucoma is the leading cause of irreversible blindness worldwide. Of the major risk factors for glaucoma (age, ocular hypertension, and genetics), only elevated intraocular pressure (IOP) is modifiable, which is largely regulated by aqueous humor outflow through the trabecular meshwork. Glucocorticoids such as dexamethasone have long been known to elevate IOP and lead to glaucoma. However, several recent studies have reported that steroid hormone estrogen levels inversely correlate with glaucoma risk, and that variants in estrogen signaling genes have been associated with glaucoma. As a result, estrogen dysregulation may contribute to glaucoma pathogenesis, and estrogen signaling may protect against glaucoma. The mechanism for estrogen-related protection against glaucoma is not completely understood but likely involves both regulation of IOP homeostasis and neuroprotection of RGCs. Based upon its known activities, estrogen signaling may promote IOP homeostasis by affecting extracellular matrix turnover, focal adhesion assembly, actin stress fiber formation, mechanosensation, and nitric oxide production. In addition, estrogen receptors in the RGCs may mediate neuroprotective functions. As a result, the estrogen signaling pathway may offer a therapeutic target for both IOP control and neuroprotection. This review examines the evidence for a relationship between estrogen and IOP and explores the possible mechanisms by which estrogen maintains IOP homeostasis.
Collapse
Affiliation(s)
- Hannah Youngblood
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Patricia V Schoenlein
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA; Department of Radiology and Georgia Cancer Center, Augusta University, Augusta, GA, USA; Department of Surgery, Augusta University, Augusta, GA, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Daniel Stamer
- Department of Ophthalmology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA; Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA.
| |
Collapse
|
22
|
Tirado-Garibay AC, Falcón-Ruiz EA, Ochoa-Zarzosa A, López-Meza JE. GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments. Int J Mol Sci 2023; 24:14993. [PMID: 37834441 PMCID: PMC10573234 DOI: 10.3390/ijms241914993] [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: 08/30/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogens and their role in cancer are well-studied, and some cancer types are classified in terms of their response to them. In recent years, a G protein-coupled estrogen receptor (GPER) has been described with relevance in cancer. GPER is a pleiotropic receptor with tissue-specific activity; in normal tissues, its activation is related to correct development and homeostasis, while in cancer cells, it can be pro- or anti-tumorigenic. Also, GPER replaces estrogen responsiveness in estrogen receptor alpha (ERα)-lacking cancer cell lines. One of the most outstanding activities of GPER is its role in epithelial-mesenchymal transition (EMT), which is relevant for metastasis development. In addition, the presence of this receptor in tumor microenvironment cells contributes to the phenotypic plasticity required for the dissemination and maintenance of tumors. These characteristics suggest that GPER could be a promising therapeutic target for regulating cancer development. This review focuses on the role of GPER in EMT in tumorigenic and associated cells, highlighting its role in relation to the main hallmarks of cancer and possible therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Joel E. López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología—FMVZ, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, Mexico; (A.C.T.-G.); (E.A.F.-R.); (A.O.-Z.)
| |
Collapse
|
23
|
Miziak P, Baran M, Błaszczak E, Przybyszewska-Podstawka A, Kałafut J, Smok-Kalwat J, Dmoszyńska-Graniczka M, Kiełbus M, Stepulak A. Estrogen Receptor Signaling in Breast Cancer. Cancers (Basel) 2023; 15:4689. [PMID: 37835383 PMCID: PMC10572081 DOI: 10.3390/cancers15194689] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogen receptor (ER) signaling is a critical regulator of cell proliferation, differentiation, and survival in breast cancer (BC) and other hormone-sensitive cancers. In this review, we explore the mechanism of ER-dependent downstream signaling in BC and the role of estrogens as growth factors necessary for cancer invasion and dissemination. The significance of the clinical implications of ER signaling in BC, including the potential of endocrine therapies that target estrogens' synthesis and ER-dependent signal transmission, such as aromatase inhibitors or selective estrogen receptor modulators, is discussed. As a consequence, the challenges associated with the resistance to these therapies resulting from acquired ER mutations and potential strategies to overcome them are the critical point for the new treatment strategies' development.
Collapse
Affiliation(s)
- Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Marzena Baran
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Alicja Przybyszewska-Podstawka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland;
| | - Magdalena Dmoszyńska-Graniczka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| |
Collapse
|
24
|
Bai J, Li Y, Yan G, Zhou J, Salmeron AG, Fategbe OT, Kumar S, Chen X, Chen DB. ICI 182,780 Attenuates Selective Upregulation of Uterine Artery Cystathionine β-Synthase Expression in Rat Pregnancy. Int J Mol Sci 2023; 24:14384. [PMID: 37762687 PMCID: PMC10532247 DOI: 10.3390/ijms241814384] [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: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Endogenous hydrogen sulfide (H2S) produced by cystathionine β-synthase (CBS) and cystathionine-γ lyase (CSE) has emerged as a novel uterine vasodilator contributing to pregnancy-associated increases in uterine blood flow, which safeguard pregnancy health. Uterine artery (UA) H2S production is stimulated via exogenous estrogen replacement and is associated with elevated endogenous estrogens during pregnancy through the selective upregulation of CBS without altering CSE. However, how endogenous estrogens regulate uterine artery CBS expression in pregnancy is unknown. This study was conducted to test a hypothesis that endogenous estrogens selectively stimulate UA CBS expression via specific estrogen receptors (ER). Treatment with E2β (0.01 to 100 nM) stimulated CBS but not CSE mRNA in organ cultures of fresh UA rings from both NP and P (gestational day 20, GD20) rats, with greater responses to all doses of E2β tested in P vs. NP UA. ER antagonist ICI 182,780 (ICI, 1 µM) completely attenuated E2β-stimulated CBS mRNA in both NP and P rat UA. Subcutaneous injection with ICI 182,780 (0.3 mg/rat) of GD19 P rats for 24 h significantly inhibited UA CBS but not mRNA expression, consistent with reduced endothelial and smooth muscle cell CBS (but not CSE) protein. ICI did not alter mesenteric and renal artery CBS and CSE mRNA. In addition, ICI decreased endothelial nitric oxide synthase mRNA in UA but not in mesenteric or renal arteries. Thus, pregnancy-augmented UA CBS/H2S production is mediated by the actions of endogenous estrogens via specific ER in pregnant rats.
Collapse
Affiliation(s)
- Jin Bai
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Yao Li
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Guofeng Yan
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Jing Zhou
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Alejandra Garcia Salmeron
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Olamide Tolulope Fategbe
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Xuejin Chen
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| |
Collapse
|
25
|
Simon AB, Derella CC, Blackburn M, Thomas J, Layman LC, Nicholson MS, Waller J, Elmarakby A, Saad KM, Harris RA. Endogenous estradiol contributes to vascular endothelial dysfunction in premenopausal women with type 1 diabetes. Cardiovasc Diabetol 2023; 22:243. [PMID: 37679748 PMCID: PMC10486136 DOI: 10.1186/s12933-023-01966-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Endogenous estrogen is cardio-protective in healthy premenopausal women. Despite this favorable action of estrogen, animal models depict a detrimental effect of estradiol on vascular function in the presence of diabetes. The present study sought to determine the role of endogenous estradiol on endothelial function in women with type 1 diabetes. METHOD 32 women with type 1 diabetes (HbA1c = 8.6 ± 1.7%) and 25 apparently healthy women (HbA1c = 5.2 ± 0.3%) participated. Flow-mediated dilation (FMD), a bioassay of nitric-oxide bioavailability and endothelial function was performed during menses (M) and the late follicular (LF) phase of the menstrual cycle to represent low and high concentrations of estrogen, respectively. In addition, a venous blood sample was collected at each visit to determine circulating concentrations of estradiol, thiobarbituric acid reactive substances (TBARS), and nitrate/nitrite (NOx), biomarkers of oxidative stress and nitric oxide, respectively. Data were collected in (1) 9 additional women with type 1 diabetes using oral hormonal birth control (HBC) (HbA1c = 8.3 ± 2.1%) during the placebo pill week and second active pill week, and (2) a subgroup of 9 demographically matched women with type 1 diabetes not using HBC (HbA1c = 8.9 ± 2.1%). RESULTS Overall, estradiol was significantly increased during the LF phase compared to M in both type 1 diabetes (Δestradiol = 75 ± 86 pg/mL) and controls (Δestradiol = 71 ± 76 pg/mL); however, an increase in TBARS was only observed in patients with type 1 diabetes (ΔTBARS = 3 ± 13 µM) compared to controls (ΔTBARS = 0 ± 4 µM). FMD was similar (p = 0.406) between groups at M. In addition, FMD increased significantly from M to the LF phase in controls (p = 0.024), whereas a decrease was observed in type 1 diabetes. FMD was greater (p = 0.015) in patients using HBC compared to those not on HBC, independent of menstrual cycle phase. CONCLUSION Endogenous estradiol increases oxidative stress and contributes to endothelial dysfunction in women with diabetes. Additionally, HBC use appears to be beneficial to endothelial function in type 1 diabetes.
Collapse
Affiliation(s)
- Abigayle B Simon
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15th Street, HS-1707, Augusta, GA, 30912, Georgia
| | - Cassandra C Derella
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15th Street, HS-1707, Augusta, GA, 30912, Georgia
| | - Marsha Blackburn
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15th Street, HS-1707, Augusta, GA, 30912, Georgia
| | - Jeffrey Thomas
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15th Street, HS-1707, Augusta, GA, 30912, Georgia
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Matthew S Nicholson
- Department of Endocrinology, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Jennifer Waller
- Department of Biostatistics and Data Science, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Ahmed Elmarakby
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Karim M Saad
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Ryan A Harris
- Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15th Street, HS-1707, Augusta, GA, 30912, Georgia.
| |
Collapse
|
26
|
Delgado Spicuzza JM, Proctor DN, Thijssen DHJ, Somani YB. Menopausal stage differences in endothelial resistance to ischemia-reperfusion injury. Physiol Rep 2023; 11:e15768. [PMID: 37734868 PMCID: PMC10513907 DOI: 10.14814/phy2.15768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND In postmenopausal women, reduced ovarian function precedes endothelial dysfunction and attenuated endothelial resistance to ischemia-reperfusion (IR) injury. We hypothesized that IR injury would lower endothelial function, with premenopausal women demonstrating the greatest protection from injury, followed by early, then late postmenopausal women. METHODS Flow-mediated dilation (FMD) was assessed at baseline and following IR injury in premenopausal (n = 11), early (n = 11; 4 ± 1.6 years since menopause), and late (n = 11; 15 ± 5.5 years since menopause) postmenopausal women. RESULTS There were significant group differences in baseline FMD (p = 0.007); post hoc analysis revealed a similar resting FMD between premenopausal (7.8% ± 2.1%) and early postmenopausal (7.1% ± 2.7%), but significantly lower FMD in late postmenopausal women (4.5% ± 2.3%). Results showed an overall decline in FMD after IR injury (p < 0.001), and a significant condition*time interaction (p = 0.048), with early postmenopausal women demonstrating the most significant decline in FMD following IR. CONCLUSION Our findings indicate that endothelial resistance to IR injury is attenuated in healthy early postmenopausal women.
Collapse
Affiliation(s)
- Jocelyn M. Delgado Spicuzza
- Integrative and Biomedical Physiology, Huck Life SciencesThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - David N. Proctor
- Integrative and Biomedical Physiology, Huck Life SciencesThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
- Kinesiology DepartmentThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Dick H. J. Thijssen
- Research Institute of Sport and Exercise ScienceLiverpool John Moores UniversityLiverpoolUK
- Radboud Institute of Health Sciences, Department of PhysiologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Yasina B. Somani
- Research Institute of Sport and Exercise ScienceLiverpool John Moores UniversityLiverpoolUK
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
27
|
Bernstein SR, Kelleher C, Khalil RA. Gender-based research underscores sex differences in biological processes, clinical disorders and pharmacological interventions. Biochem Pharmacol 2023; 215:115737. [PMID: 37549793 PMCID: PMC10587961 DOI: 10.1016/j.bcp.2023.115737] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Earlier research has presumed that the male and female biology is similar in most organs except the reproductive system, leading to major misconceptions in research interpretations and clinical implications, with serious disorders being overlooked or misdiagnosed. Careful research has now identified sex differences in the cardiovascular, renal, endocrine, gastrointestinal, immune, nervous, and musculoskeletal systems. Also, several cardiovascular, immunological, and neurological disorders have shown differences in prevalence and severity between males and females. Genetic variations in the sex chromosomes have been implicated in several disorders at young age and before puberty. The levels of the gonadal hormones estrogen, progesterone and testosterone and their receptors play a role in the sex differences between adult males and premenopausal women. Hormonal deficiencies and cell senescence have been implicated in differences between postmenopausal and premenopausal women. Specifically, cardiovascular disorders are more common in adult men vs premenopausal women, but the trend is reversed with age with the incidence being greater in postmenopausal women than age-matched men. Gender-specific disorders in females such as polycystic ovary syndrome, hypertension-in-pregnancy and gestational diabetes have attained further research recognition. Other gender-related research areas include menopausal hormone therapy, the "Estrogen Paradox" in pulmonary arterial hypertension being more predominant but less severe in young females, and how testosterone may cause deleterious effects in the kidney while having vasodilator effects in the coronary circulation. This has prompted the National Institutes of Health (NIH) initiative to consider sex as a biological variable in research. The NIH and other funding agencies have provided resources to establish state-of-the-art centers for women health and sex differences in biology and disease in several academic institutions. Scientific societies and journals have taken similar steps to organize specialized conferences and publish special issues on gender-based research. These combined efforts should promote research to enhance our understanding of the sex differences in biological systems beyond just the reproductive system, and provide better guidance and pharmacological tools for the management of various clinical disorders in a gender-specific manner.
Collapse
Affiliation(s)
- Sofia R Bernstein
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Caroline Kelleher
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Mott MN, Goeders NE. Methamphetamine-induced vaginal lubrication in rats. J Sex Med 2023; 20:1145-1152. [PMID: 37291060 DOI: 10.1093/jsxmed/qdad076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Based on previous studies of vaginal lubrication as well as our own previously reported interview study of women who self-reported methamphetamine (meth)-induced vaginal lubrication, in the current study we sought to determine the potential dose-response relationship leading to meth-induced vaginal lubrication. We also developed an animal model to study the reported effects and examine potential mechanisms mediating this phenomenon. AIM We sought to characterize the effects of meth on vaginal lubrication in an animal model with the aim of providing a potential framework for new mechanisms that incorporate novel therapeutic agents for the treatment of vaginal dryness. METHODS Vaginal lubrication was measured via insertion of a preweighed, cotton-tipped swab into the vaginal canal of anesthetized rats following treatment with various doses of intravenous (IV) meth, up to 0.96 mg/kg, and after additional pharmacological manipulations, including administration of a nitric oxide synthase inhibitor and an estrogen receptor antagonist. Plasma signaling molecules, including estradiol, progesterone, testosterone, nitric oxide, and vasoactive intestinal polypeptide, were measured immediately before and at 9 time points after IV meth administration. Blood was collected via a previously implanted chronic indwelling jugular catheter and analyzed by use of commercially available kits per the manufacturer's instructions. OUTCOMES Outcomes for this study include the measurement of vaginal lubrication in anesthetized rats following various pharmacological manipulations and plasma levels of various signaling molecules. RESULTS Meth dose-dependently increased vaginal lubrication in anesthetized female rats. Meth significantly increased plasma levels compared to baseline of estradiol (2 and 15 minutes after meth infusion) as well as progesterone, testosterone, and nitric oxide (10 minutes after meth infusion). Also, vasoactive intestinal polypeptide decreased significantly compared to baseline for 45 minutes following meth infusion. Our data further suggest that nitric oxide, but not estradiol, is critical in the production of vaginal secretions in response to meth. CLINICAL IMPLICATIONS This study has far-reaching implications for women who are suffering from vaginal dryness and for whom estrogen therapy is unsuccessful, as the investigation has demonstrated that meth presents a novel mechanism for producing vaginal lubrication that can be targeted pharmacologically. STRENGTHS AND LIMITATIONS This study is, to our knowledge, the first performed to measure the physiological sexual effects of meth in an animal model. Animals were anesthetized when they were administered meth. In an ideal situation, animals would be self-administering the drug to recapitulate better the contingent nature of drug taking; however, this method was not feasible for the study reported here. CONCLUSION Methamphetamine increases vaginal lubrication in female rats through a nitric oxide-dependent mechanism.
Collapse
Affiliation(s)
- Maggie N Mott
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Shreveport, LA, United States
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
- Louisiana Addiction Research Center, Shreveport, LA, United States
| |
Collapse
|
29
|
Priyadarshini E, Parambil AM, Rajamani P, Ponnusamy VK, Chen YH. Exposure, toxicological mechanism of endocrine disrupting compounds and future direction of identification using nano-architectonics. ENVIRONMENTAL RESEARCH 2023; 225:115577. [PMID: 36871939 DOI: 10.1016/j.envres.2023.115577] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Endocrine-disrupting compounds (EDC) are a group of exogenous chemicals that structurally mimic hormones and interfere with the hormonal signaling cascade. EDC interacts with hormone receptors, transcriptional activators, and co-activators, altering the signaling pathway at both genomic and non-genomic levels. Consequently, these compounds are responsible for adverse health ailments such as cancer, reproductive issues, obesity, and cardiovascular and neurological disorders. The persistent nature and increasing incidence of environmental contamination from anthropogenic and industrial effluents have become a global concern, resulting in a movement in both developed and developing countries to identify and estimate the degree of exposure to EDC. The U.S. Environment Protection Agency (EPA) has outlined a series of in vitro and in vivo assays to screen potential endocrine disruptors. However, the multidisciplinary nature and concerns over the widespread application demand alternative and practical techniques for identifying and estimating EDC. The review chronicles the state-of-art 20 years (1990-2023) of scientific literature regarding EDC's exposure and molecular mechanism, highlighting the toxicological effects on the biological system. Alteration in signaling mechanisms by representative endocrine disruptors such as bisphenol A (BPA), diethylstilbestrol (DES), and genistein has been emphasized. We further discuss the currently available assays and techniques for in vitro detection and propose the prominence of designing nano-architectonic-sensor substrates for on-site detection of EDC in the contaminated aqueous environment.
Collapse
Affiliation(s)
- Eepsita Priyadarshini
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ajith Manayil Parambil
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Vinoth Kumar Ponnusamy
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, Taiwan; Department of Chemistry, National Sun Yat-sen University (NSYSU), Kaohsiung City, 804, Taiwan; PhD Program in Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, 811, Taiwan.
| | - Yi-Hsun Chen
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
30
|
Berbrier DE, Leone CA, Adler TE, Bender JR, Taylor HS, Stachenfeld NS, Usselman CW. Effects of androgen excess and body mass index on endothelial function in women with polycystic ovary syndrome. J Appl Physiol (1985) 2023; 134:868-878. [PMID: 36861670 DOI: 10.1152/japplphysiol.00583.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with endothelial dysfunction; whether this is attributable to comorbid hyperandrogenism and/or obesity remains to be established. Therefore, we 1) compared endothelial function between lean and overweight/obese (OW/OB) women with and without androgen excess (AE)-PCOS and 2) examined androgens as potential modulators of endothelial function in these women. The flow-mediated dilation (FMD) test was applied in 14 women with AE-PCOS (lean: n = 7; OW/OB: n = 7) and 14 controls (CTRL; lean: n = 7, OW/OB: n = 7) at baseline (BSL) and following 7 days of ethinyl estradiol supplementation (EE; 30 µg/day) to assess the effect of a vasodilatory therapeutic on endothelial function; at each time point we assessed peak increases in diameter during reactive hyperemia (%FMD), shear rate, and low flow-mediated constriction (%LFMC). BSL %FMD was attenuated in lean AE-PCOS versus both lean CTRL (5.2 ± 1.5 vs. 10.3 ± 2.6%, P < 0.01) and OW/OB AE-PCOS (5.2 ± 1.5 vs. 6.6 ± 0.9%, P = 0.048). A negative correlation between BSL %FMD and free testosterone was observed in lean AE-PCOS only (R2 = 0.68, P = 0.02). EE increased %FMD in both OW/OB groups (CTRL: 7.6 ± 0.6 vs. 10.4 ± 2.5%, AE-PCOS: 6.6 ± 0.9 vs. 9.6 ± 1.7%, P < 0.01), had no impact on %FMD in lean AE-PCOS (5.17 ± 1.5 vs. 5.17 ± 1.1%, P = 0.99), and reduced %FMD in lean CTRL (10.3 ± 2.6 vs. 7.6 ± 1.2%, P = 0.03). Collectively, these data indicate that lean women with AE-PCOS exhibit more severe endothelial dysfunction than their OW/OB counterparts. Furthermore, endothelial dysfunction appears to be mediated by circulating androgens in lean but not in OW/OB AE-PCOS, suggesting a difference in the endothelial pathophysiology of AE-PCOS between these phenotypes.NEW & NOTEWORTHY We present evidence for marked endothelial dysfunction in lean women with androgen excess polycystic ovary syndrome (AE-PCOS) that is 1) associated with free testosterone levels, 2) impaired relative to overweight/obese women with AE-PCOS, and 3) unchanged following short-term ethinyl estradiol supplementation. These data indicate an important direct effect of androgens on the vascular system in women with AE-PCOS. Our data also suggest that the relationship between androgens and vascular health differs between phenotypes of AE-PCOS.
Collapse
Affiliation(s)
- Danielle E Berbrier
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Cheryl A Leone
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
| | - Tessa E Adler
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
| | - Jeffrey R Bender
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut, United States
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Nina S Stachenfeld
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Charlotte W Usselman
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
- McGill Research Centre for Physical Activity and Health, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Zhou K, Xiao J, Wang H, Ni B, Huang J, Long X. Estradiol regulates oxidative stress and angiogenesis of myocardial microvascular endothelial cells via the CDK1/CDK2 pathway. Heliyon 2023; 9:e14305. [PMID: 36942258 PMCID: PMC10023923 DOI: 10.1016/j.heliyon.2023.e14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Cardiovascular diseases remain the leading cause of death, morbidity, and disability. Recently, it has been reported that gonadal hormones such as estradiol can act on membrane receptors and activate intracellular signaling mechanisms, thereby altering cellular function. This study aims to explore the function and molecular mechanism of estradiol on cardiac microvascular endothelial cells (CMVECs). Estradiol had low toxicity to CMVECs. Hypoxia/reoxygenation (H/R) stimulation inhibited the proliferation and migration of CMVECs, while estradiol significantly promoted proliferation and migration. Estradiol inhibited il-1, IL6, and TNF-α secretion levels after H/R stimulation. Meanwhile, estradiol inhibits oxidative stress and promotes angiogenesis. Further, estradiol upregulated the gene and protein levels of cyclin-dependent kinases 1 (CDK1) and CDK2 after H/R stimulation. When knocking down CDK1 and CDK2 of CMVECs, estradiol did not affect the protein expression of Cyclin E1 and Cyclin D1. Meanwhile, the regulatory effect of estradiol on oxidative stress, angiogenesis, and inflammatory response was significantly weakened or even disappeared. In conclusion, estradiol mediates oxidative stress and angiogenesis of myocardial microvascular endothelial cells by regulating the CDK/cyclin signaling pathway.
Collapse
Affiliation(s)
- Ke Zhou
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Jun Xiao
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
- Corresponding author.
| | - Hao Wang
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Bing Ni
- Institute of Immunology of Army Medical University, Chongqing, 400014, China
| | - Jietao Huang
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Xueyuan Long
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| |
Collapse
|
32
|
Guthrie GL, Almutlaq RN, Sugahara S, Butt MK, Brooks CR, Pollock DM, Gohar EY. G protein-coupled estrogen receptor 1 regulates renal endothelin-1 signaling system in a sex-specific manner. Front Physiol 2023; 14:1086973. [PMID: 36733911 PMCID: PMC9887121 DOI: 10.3389/fphys.2023.1086973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Demographic studies reveal lower prevalence of hypertension among premenopausal females compared to age-matched males. The kidney plays a central role in the maintenance of sodium (Na+) homeostasis and consequently blood pressure. Renal endothelin-1 (ET-1) is a pro-natriuretic peptide that contributes to sex differences in blood pressure regulation and Na+ homeostasis. We recently showed that activation of renal medullary G protein-coupled estrogen receptor 1 (GPER1) promotes ET-1-dependent natriuresis in female, but not male, rats. We hypothesized that GPER1 upregulates the renal ET-1 signaling system in females, but not males. To test our hypothesis, we determined the effect of GPER1 deletion on ET-1 and its downstream effectors in the renal cortex, outer and inner medulla obtained from 12-16-week-old female and male mice. GPER1 knockout (KO) mice and wildtype (WT) littermates were implanted with telemetry transmitters for blood pressure assessment, and we used metabolic cages to determine urinary Na+ excretion. GPER1 deletion did not significantly affect 24-h mean arterial pressure (MAP) nor urinary Na+ excretion. However, GPER1 deletion decreased urinary ET-1 excretion in females but not males. Of note, female WT mice had greater urinary ET-1 excretion than male WT littermates, whereas no sex differences were observed in GPER1 KO mice. GPER1 deletion increased inner medullary ET-1 peptide content in both sexes but increased outer medullary ET-1 content in females only. Cortical ET-1 content increased in response to GPER1 deletion in both sexes. Furthermore, GPER1 deletion notably increased inner medullary ET receptor A (ETA) and decreased outer medullary ET receptor B (ETB) mRNA expression in male, but not female, mice. We conclude that GPER1 is required for greater ET-1 excretion in females. Our data suggest that GPER1 is an upstream regulator of renal medullary ET-1 production and ET receptor expression in a sex-specific manner. Overall, our study identifies the role of GPER1 as a sex-specific upstream regulator of the renal ET-1 system.
Collapse
Affiliation(s)
- Ginger L. Guthrie
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rawan N. Almutlaq
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sho Sugahara
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Maryam K. Butt
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Craig R. Brooks
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David M. Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eman Y. Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States,*Correspondence: Eman Y. Gohar,
| |
Collapse
|
33
|
Tissue Derivation and Biological Sex Uniquely Mediate Endothelial Cell Protein Expression, Redox Status, and Nitric Oxide Synthesis. Cells 2022; 12:cells12010093. [PMID: 36611888 PMCID: PMC9818567 DOI: 10.3390/cells12010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Human endothelial cells are routinely utilized in cardiovascular research to provide a translational foundation for understanding how the vascular endothelium functions in vivo. However, little attention has been given to whether there are sex specific responses in vitro. Similarly, it is unclear whether endothelial cells derived from distinct tissues behave in a homogenous manner. Herein, we demonstrate that marked sex differences exist within, and between, commonly utilized human primary endothelial cells from healthy donors, with respect to redox status, nitric oxide synthesis, and associated proteins that can mediate their expression. Further, we demonstrate that endothelial cells respond uniquely to inflammatory insult in a sex- and tissue origin-dependent manner. Our findings suggest sex and tissue derivation may need to be considered when studying endothelial cells in vitro as cells derived from distinct tissue and sexes may not behave interchangeably.
Collapse
|
34
|
Azizian H, Farhadi Z, Khaksari M. Selective estrogen receptor α and β antagonist aggravate cardiovascular dysfunction in type 2 diabetic ovariectomized female rats. Horm Mol Biol Clin Investig 2022; 43:427-436. [PMID: 35512107 DOI: 10.1515/hmbci-2021-0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/25/2022] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Type 2 diabetes (T2D) is a major risk factor for cardiovascular disorders (CVD), characterized by pathological diastolic as well as systolic dysfunction, ventricular dilation, and cardiomyocyte hypertrophy. CVD is the main cause of death in postmenopausal women. Estradiol (E2) has protective effects on cardiovascular function. The biological effects of E2 are mainly mediated by classical estrogen receptors (ERs). The present study aimed to investigate the cardioprotective effects of classical ERs in ovariectomized (OVX) diabetic female rats. METHODS T2D was induced in female rats by high-fat diet feeding along with a low dose of streptozotocin. Then diabetic animals were divided into eight groups: Sham-control, OVX, OVX + Vehicle (Veh), OVX + E2, OVX + E2 + MPP (ERα antagonist), OVX + E2 + PHTPP (ERβ antagonist), OVX + E2 + Veh, OVX + E2 + MPP + PHTPP. Animals received E2, MPP, and PHTPP every four days for 28 days. At the end blood was collected, serum separated, and used for biochemical parameters. Heart tissue was used for cardiac angiotensin II and cytokines measurement. RESULTS E2 treatment improved the metabolic disorders caused by T2D, and its receptor antagonists intensified the effects of T2D on the metabolic status. Also, E2 therapy decreased cardiac inflammatory cytokines, and MPP and PHTPP increased cardiac inflammation by increasing TNF-α and IL-6 and decreasing IL-10. CONCLUSIONS Classical ERs have protective effects on diabetic hearts by improving the metabolic status and inflammatory balance.
Collapse
Affiliation(s)
- Hossein Azizian
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeinab Farhadi
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
35
|
Erewele EO, Castellon M, Loya O, Marshboom G, Schwartz A, Yerlioglu K, Callahan C, Chen J, Minshall RD, Oliveira SD. Hypoxia-induced pulmonary hypertension upregulates eNOS and TGF-β contributing to sex-linked differences in BMPR2 +/R899X mutant mice. Pulm Circ 2022; 12:e12163. [PMID: 36484056 PMCID: PMC9722973 DOI: 10.1002/pul2.12163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/28/2022] Open
Abstract
Dysfunctional bone morphogenetic protein receptor 2 (BMPR2) and endothelial nitric oxide synthase (eNOS) have been largely implicated in the pathogenesis of pulmonary arterial hypertension (PAH); a life-threatening cardiopulmonary disease. Although the incident of PAH is about three times higher in females, males with PAH usually have a worse prognosis, which seems to be dependent on estrogen-associated cardiac and vascular protection. Here, we evaluated whether hypoxia-induced pulmonary hypertension (PH) in humanized BMPR2+/R899X loss-of-function mutant mice contributes to sex-associated differences observed in PAH by altering eNOS expression and inducing expansion of hyperactivated TGF-β-producing pulmonary myofibroblasts. To test this hypothesis, male and female wild-type (WT) and BMPR2+/R899X mutant mice were kept under hypoxic or normoxic conditions for 4 weeks, and then right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH) were measured. Chronic hypoxia exposure elevated RVSP, inducing RVH in both groups, with a greater effect in BMPR2+/R899X female mice. Lung histology revealed no differences in vessel thickness/area between sexes, suggesting RVSP differences in this model are unlikely to be in response to sex-dependent vascular narrowing. On the other hand, hypoxia exposure increased vascular collagen deposition, the number of TGF-β-associated α-SMA-positive microvessels, and eNOS expression, whereas it also reduced caveolin-1 expression in the lungs of BMPR2+/R899X females compared to males. Taken together, this brief report reveals elevated myofibroblast-derived TGF-β and eNOS-derived oxidants contribute to pulmonary microvascular muscularization and sex-linked differences in incidence, severity, and outcome of PAH.
Collapse
Affiliation(s)
- Ejehi O Erewele
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Maricela Castellon
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
- Cardiovascular Research Center, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Omar Loya
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Glenn Marshboom
- Department of Pharmacology and Regenerative Medicine, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Andrew Schwartz
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Kayla Yerlioglu
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Christopher Callahan
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Jiwang Chen
- Cardiovascular Research Center, College of Medicine University of Illinois at Chicago Chicago Illinois USA
- Department of Medicine, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Richard D Minshall
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
- Department of Pharmacology and Regenerative Medicine, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| | - Suellen D Oliveira
- Department of Anesthesiology, College of Medicine University of Illinois at Chicago Chicago Illinois USA
- Department of Physiology & Biophysics, College of Medicine University of Illinois at Chicago Chicago Illinois USA
| |
Collapse
|
36
|
Sex-based differences in conjunctival goblet cell responses to pro-inflammatory and pro-resolving mediators. Sci Rep 2022; 12:16305. [PMID: 36175572 PMCID: PMC9522727 DOI: 10.1038/s41598-022-20177-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
Many conjunctival inflammatory diseases differ between the sexes and altered conjunctival goblet cells (CGCs) response is often involved. Inflammation is initiated by the release of pro-inflammatory mediators and terminated by the biosynthesis of specialized pro-resolution mediators (SPMs). Herein, we determined the sex-based difference in the responses of CGCs to inflammatory stimuli or pro-resolving lipid SPMs and their interaction with sex hormones. GCs were cultured from pieces of human conjunctiva in RPMI media. CGCs were transferred 24 h before the start of experiments to phenol red-free and FBS-free media to minimize exogenous hormones. RT-PCR, immunofluorescence microscopy (IF), and Western Blot (WB) were performed to determine the presence of sex hormone receptors. Cellular response to pro-inflammatory stimuli or SPMs was studied by measuring the increase in intracellular [Ca2+] ([Ca2+]i) using fura 2/AM microscopy. Use of RT-PCR demonstrated estrogen receptor (ER) α in 4/5 males and 3/3 females; ERβ in 2/4 males and 2/3 females; and androgen receptors (AR) in 3/3 male and 3/3 female CGCs. Positive immunoreactivity by IF and protein expression by WB was detected using antibodies for the ERα and ERβ in 3/3 males and 3/3 females, while AR were only present in males. Significantly different Ca2+ responses between sexes were found with carbachol only at 10–3 M, but not with histamine or leukotriene (LT) B4 at any concentration used. Incubation with dihydrotestosterone (DHT), estrone (E1), or estradiol (E2) at 10–7 M for 30 min significantly inhibited the LTB4-stimulated [Ca2+]i increase in male and female CGCs. Incubation with DHT, E1, and E2 overnight significantly inhibited the LTB4 response in females, while DHT and E2 significantly inhibited the LTB4 response in males. The SPM lipoxin A4 (LXA4) (10–9–10−8 M), but not the resolvins D1 or D2, induced an [Ca2+]i increase that was significantly higher in males compared to females. We conclude that male and female CGCs showed differences in the expression of sex hormone receptors. Treatment with sex hormones altered pro-inflammatory mediator LTB4-induced response. Males compared to females have a higher response to the ω-6-fatty acid derived SPM LXA4, indicating males may terminate inflammation in conjunctival goblet cells faster than females.
Collapse
|
37
|
Chen L, Hu Z, Wang X, Song Y, Chen Z, Zhang L, Zheng C, Vallis J, Zhou H, Cao X, Tian Y, Cai J, Gu R, Huang Y, Wang Z. Age at Menarche and Menopause, Reproductive Lifespan, and Risk of Cardiovascular Events Among Chinese Postmenopausal Women: Results From a Large National Representative Cohort Study. Front Cardiovasc Med 2022; 9:870360. [PMID: 36158833 PMCID: PMC9500155 DOI: 10.3389/fcvm.2022.870360] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background At present, the association between age at menarche and menopause, reproductive lifespan, and cardiovascular disease (CVD) risk among Chinese postmenopausal women is not clear, and some related researches are contradictory. Methods A total of 6,198 Chinese postmenopausal women with a mean age of 63.6 years were enrolled at baseline in 2012-2015 and followed up for 5 years. A standardized questionnaire was used to collect relevant information by well-trained interviewers. Physical examination of the participants was performed by trained medical staff. CVD events were observed during follow-up. Cox proportional hazards models were used to estimate hazard ratios between reproductive characteristics and CVD events. Results Age at menarche was positively associated with CVD events (HR, 1.106; 95%CI, 1.047-1.167). There was a negative association between age at menopause and CVD risk in postmenopausal women with comorbidity (HR, 0.952; 95%CI, 0.909-0.996). Reproductive lifespan was negatively associated with CVD events (HR, 0.938; 95%CI, 0.880-0.999). The CVD risk increased by 10.6% for every 1-year increase in age at menarche. The CVD risk reduced by 6.2% for every 1-year increase in age at menopause in women with comorbidity. The CVD risk reduced by 3.8% for every 1-year increase in reproductive lifespan. Conclusions Based on the large prospective study with a nationally representative sample, Chinese postmenopausal women with late age at menarche and shorter reproductive lifespan have higher risk of CVD events.
Collapse
Affiliation(s)
- Lu Chen
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhen Hu
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Wang
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuxin Song
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zuo Chen
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Linfeng Zhang
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Congyi Zheng
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jillian Vallis
- PGY3 General Surgery, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Haoqi Zhou
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Cao
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yixin Tian
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiayin Cai
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Runqing Gu
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Huang
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zengwu Wang
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, National Clinical Research Center of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
38
|
Liu XY, Li P, Li XS, Simoncini T, Cheng Y. 17β-Estradiol nongenomically induces vasodilation is enhanced by promoting phosphorylation of endophilin A2. Gynecol Endocrinol 2022; 38:644-650. [PMID: 35730594 DOI: 10.1080/09513590.2022.2088731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/22/2022] [Accepted: 06/08/2022] [Indexed: 11/11/2022] Open
Abstract
ObjectiveA previous study found that the tyrosine phosphorylation of endophilin A2 (Endo II) was responsible for increase surface expression of MT1-MMP and ECM degradation; however, there is little information about whether Endo II could influence membrane estrogen receptors (mERs) and its functions.Materials and methodsIn the present study, Human umbilical vein endothelial cells (HUVECs) were treated with E2, PPT, DPN, ICI 182780, Endo siRNA or negative control siRNA, and the biological behavior of the treated cells was observed. The mice were randomly divided into AAV-control-shRNA + Ach, AAV-Endo II-shRNA + Ach, AAV-control-shRNA + E2, AAV-Endo II-shRNA + E2 groups and the thoracic aorta were isolated, cut into 2-mm rings, then the wall tension was detected.ResultsWe found that 17β-Estradiol (E2) enhanced mERα protein level, which was further increased after knocking down Endo II, the mechanism maybe involved in E2-induced tyrosine phosphorylation of Endo II. In addition, we also observed that Endo II blocked the activation of Akt, ERK1/2 and eNOS signaling in HUVECs treated with E2. E2 induced vasodilation was significantly increased by silencing of Endo II expression.ConclusionOur study provided a sound basis to selective modulate Endo II for E2's nongenomic pathway, which can be benefit for cardiovascular system.
Collapse
Affiliation(s)
- Xiao-Yun Liu
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Clinical Pharmacy (School of Integrative Pharmacy, Institute of Integrative Pharmaceutical Research), Guangdong Pharmaceutical University, Guangzhou, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ping Li
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Sa Li
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Yang Cheng
- Department of Gynecology and Obstetrics, Guangzhou First People's Hospital, South China University of Technology, Guang zhou, Guangdong, China
| |
Collapse
|
39
|
Bechmann N, Calsina B, Richter S, Pietzsch J. Therapeutic Potential of Nitric Oxide‒Releasing Selective Estrogen Receptor Modulators in Malignant Melanoma. J Invest Dermatol 2022; 142:2217-2227. [PMID: 34990694 DOI: 10.1016/j.jid.2021.12.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 10/24/2022]
Abstract
Malignant melanoma has a steadily increasing incidence, but treatment options are still limited, and the prognosis for patients, especially for men, is poor. To investigate whether targeting estrogen receptor (ER) signaling is a valid therapeutic approach, we retrospectively analyzed ER gene expression profiles in 448 patients with melanoma. High ERα gene (ESR1) expression was associated with improved overall survival (hazard ratio = 0.881; 95% confidence interval = 0.793-0.979; P = 0.018) and increased with tumor stage, whereas ERβ gene (ESR2) expression did not change with tumor progression. This seemingly protective function of ERα led us to speculate that specific targeting of ERβ has a therapeutic benefit in malignant melanoma. An ERβ-selective ER modulator with nitric oxide‒releasing moiety (nitric oxide‒releasing selective ER modulator 4d [NO-SERM 4d]) significantly reduced the prometastatic behavior of two melanoma cell lines (A2058 and MEL-JUSO). Epithelial‒mesenchymal transition in melanoma is consistent with a switch from E- to N-cadherin expression, mediating the invasive phenotype. NO-SERM 4d reduced N-cadherin expression and impaired spheroid formation in A2058 cells. In addition, the growth of A2058 spheroids was significantly reduced, confirming the antitumorigenic potential of NO-SERM 4d. Targeting ERβ signaling combined with targeted nitric oxide release represents a promising therapeutic approach in malignant melanoma that has the potential to prevent metastatic spread and reduce tumor growth.
Collapse
Affiliation(s)
- Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, School of Medicine, Technische Universität Dresden, Dresden, Germany; Department of Medicine III, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Experimental Diabetology (DIAB), German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Hereditary Endocrine Cancer Group, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, School of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum, Dresden-Rossendorf, Dresden, Germany; Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
40
|
Associations between Age at Menarche and Dietary Patterns with Blood Pressure in Southwestern Chinese Adults. Nutrients 2022; 14:nu14081610. [PMID: 35458171 PMCID: PMC9029714 DOI: 10.3390/nu14081610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/05/2022] [Accepted: 04/09/2022] [Indexed: 11/17/2022] Open
Abstract
The aim of our study was to examine the relationship between age at menarche (AM) and hypertension and to evaluate whether different dietary patterns have an effect on associations between AM and hypertension in a large-scale Han Chinese population in southwest China. A cross-sectional study was performed that included 44,900 participants from 17 districts in southwest China from September 2018 to January 2019. The study comprised 23,805 individuals in the final analysis. Logistic regression and multivariable linear regression were applied to estimate the dietary pattern-specific associations between AM and hypertension or systolic/diastolic blood pressure (SBP/DBP). Restricted cubic spline regression was utilized to calculate the shape of the relationship between AM and the odds ratio of hypertension. After adjusting for multiple variables, women who had a history of AM > 14 years were associated with an increased risk of hypertension (OR 1.12, 95%CI 1.04−1.19) and elevated levels of SBP (β 0.90, 95%CI 0.41−1.38) compared with those with AM ≤ 14 years among the total population, and this association was still statistically significant when we further adjusted for body mass index (BMI). In participants with AM > 14 years, the odds ratio values of hypertension increased with increasing menarche age. After stratification by age at recruitment, the positive association between menarche age and hypertension only remained in the middle-aged group, and this association was not found in the young and old groups. After stratification by the Dietary Approaches to Stop Hypertension (DASH) score among the total population, the positive association between AM and hypertension was presented only in the low DASH score group; however, this association was not found in the high DASH score group. Women who have a history of AM > 14 years should pay close attention to blood pressure levels and incorporate the DASH diet more in order to achieve the early prevention of hypertension, especially middle-aged women.
Collapse
|
41
|
Wu J, Fang S, Lu KT, Kumar G, Reho JJ, Brozoski DT, Otanwa AJ, Hu C, Nair AR, Wackman KK, Agbor LN, Grobe JL, Sigmund CD. Endothelial Cullin3 Mutation Impairs Nitric Oxide-Mediated Vasodilation and Promotes Salt-Induced Hypertension. FUNCTION (OXFORD, ENGLAND) 2022; 3:zqac017. [PMID: 35493997 PMCID: PMC9045850 DOI: 10.1093/function/zqac017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/13/2023]
Abstract
Human hypertension caused by in-frame deletion of CULLIN3 exon-9 (Cul3∆9) is driven by renal and vascular mechanisms. We bred conditionally activatable Cul3∆9 transgenic mice with tamoxifen-inducible Tie2-CREERT2 mice to test the importance of endothelial Cul3. The resultant mice (E-Cul3∆9) trended towards elevated nighttime blood pressure (BP) correlated with increased nighttime activity, but displayed no difference in daytime BP or activity. Male and female E-Cul3∆9 mice together exhibited a decline in endothelial-dependent relaxation in carotid artery. Male but not female E-Cul3∆9 mice displayed severe endothelial dysfunction in cerebral basilar artery. There was no impairment in mesenteric artery and no difference in smooth muscle function, suggesting the effects of Cul3∆9 are arterial bed-specific and sex-dependent. Expression of Cul3∆9 in primary mouse aortic endothelial cells decreased endogenous Cul3 protein, phosphorylated (S1177) endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production. Protein phosphatase (PP) 2A, a known Cul3 substrate, dephosphorylates eNOS. Cul3∆9-induced impairment of eNOS activity was rescued by a selective PP2A inhibitor okadaic acid, but not by a PP1 inhibitor tautomycetin. Because NO deficiency contributes to salt-induced hypertension, we tested the salt-sensitivity of E-Cul3∆9 mice. While both male and female E-Cul3∆9 mice developed salt-induced hypertension and renal injury, the pressor effect of salt was greater in female mutants. The increased salt-sensitivity in female E-Cul3∆9 mice was associated with decreased renovascular relaxation and impaired natriuresis in response to a sodium load. Thus, CUL3 mutations in the endothelium may contribute to human hypertension in part through decreased endothelial NO bioavailability, renovascular dysfunction, and increased salt-sensitivity of BP.
Collapse
Affiliation(s)
- Jing Wu
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Shi Fang
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA,Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Ko-Ting Lu
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Gaurav Kumar
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - John J Reho
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Daniel T Brozoski
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Adokole J Otanwa
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Chunyan Hu
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Anand R Nair
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Kelsey K Wackman
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Larry N Agbor
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Justin L Grobe
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | | |
Collapse
|
42
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
43
|
Goyal A, Agrawal N, Jain A, Gupta JK, Garabadu D. Role of caveolin-eNOS platform and mitochondrial ATP-sensitive potassium channel in abrogated cardioprotective effect of ischemic preconditioning in postmenopausal women. BRAZ J PHARM SCI 2022; 58. [DOI: 10.1590/s2175-97902022e20081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
| | | | - Ankit Jain
- Dr. Hari Singh Gour Central University, India
| | | | | |
Collapse
|
44
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
45
|
D'Agata MN, Hoopes EK, Berube FR, Hirt AE, Kuczmarski AV, Ranadive SM, Wenner MM, Witman MA. Evidence of reduced peripheral microvascular function in young Black women across the menstrual cycle. J Appl Physiol (1985) 2021; 131:1783-1791. [PMID: 34709068 PMCID: PMC8714980 DOI: 10.1152/japplphysiol.00452.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022] Open
Abstract
Black women (BLW) have a higher prevalence of cardiovascular disease (CVD) morbidity and mortality compared with White women (WHW). A racial disparity in CVD risk has been identified early in life as young adult BLW demonstrate attenuated vascular function compared with WHW. Previous studies comparing vascular function between premenopausal WHW and BLW have been limited to the early follicular (EF) phase of the menstrual cycle, which may not reflect their vascular function during other menstrual phases. Therefore, we evaluated peripheral microvascular function in premenopausal WHW and BLW using passive leg movement (PLM) during three menstrual phases: EF, ovulation (OV), and mid-luteal (ML). We hypothesized that microvascular function would be augmented during the OV and ML phases compared with the EF phase in both groups, but would be attenuated in BLW compared with WHW at all three phases. PLM was performed on 26 apparently healthy premenopausal women not using hormonal contraceptives: 15 WHW (23 ± 3 yr), 11 BLW (24 ± 5 yr). There was a main effect of race on the overall change in leg blood flow (ΔLBF) (P = 0.01) and leg blood flow area under the curve (LBF AUC) (P = 0.02), such that LBF was lower in BLW. However, there was no effect of phase on ΔLBF (P = 0.69) or LBF AUC (P = 0.65), nor an interaction between race and phase on ΔLBF (P = 0.37) or LBF AUC (P = 0.75). Despite peripheral microvascular function being unchanged across the menstrual cycle, a racial disparity was apparent as microvascular function was attenuated in BLW compared with WHW across the menstrual cycle.NEW & NOTEWORTHY This is the first study to compare peripheral microvascular function between young, otherwise healthy Black women and White women at multiple phases of the menstrual cycle. Our novel findings demonstrate a significant effect of race on peripheral microvascular function such that Black women exhibit significant attenuations in microvascular function across the menstrual cycle compared with White women.
Collapse
Affiliation(s)
- Michele N D'Agata
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Elissa K Hoopes
- Department of Behavioral Health and Nutrition, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Felicia R Berube
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Alexandra E Hirt
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Andrew V Kuczmarski
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Sushant M Ranadive
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| | - Melissa A Witman
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
46
|
McIntosh RC, Lobo JD, Yang A, Schneiderman N. Brainstem network connectivity with mid-anterior insula predicts lower systolic blood pressure at rest in older adults with hypertension. J Hum Hypertens 2021; 35:1098-1108. [PMID: 33462388 PMCID: PMC8919345 DOI: 10.1038/s41371-020-00476-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/13/2020] [Accepted: 12/10/2020] [Indexed: 11/09/2022]
Abstract
Central regulation of heart rate and blood pressure provides the bases for a neurogenic mechanism of hypertension (HTN). Post menopause (PM) age coincides with changes in resting state functional brain connectivity (rsFC) as well as increased risk for HTN. Whether the neural networks underpinning cardioautonomic control differ between PM women with and without HTN is unclear. Phenotypic and functional neuroimaging data from the Nathan Kline Institute was first evaluated for group differences in intrinsic network connectivity between 22 HTN post menopausal women and 22 normotensive controls. Intrinsic rsFC of the midbrain-brainstem-cerebellar network with bilateral mid-anterior insula was lower in women with HTN (FWE-corrected, p < 0.05). Z-scores indicating rsFC of these regions were extracted from the 44 PM women and a cohort of 111 adults, not presenting with metabolic or neurodegenerative disease, and compared to in-office systolic and diastolic blood pressure. Lower rsFC of the left (r = -0.17, p = 0.019) and right (r = -0.14, p = 0.048) mid-anterior insula with brainstem nuclei was associated with higher systolic blood pressure in the combined sample. The magnitude of this effect in men and women of post menopausal age supports a neurogenic mechanism for blood pressure regulation in older adults with HTN.
Collapse
Affiliation(s)
- Roger C McIntosh
- Department of Psychology, University of Miami, Coral Gables, FL, USA.
| | - Judith D Lobo
- Department of Psychology, University of Miami, Coral Gables, FL, USA
| | - Anting Yang
- Department of Psychology, University of Miami, Coral Gables, FL, USA
| | - Neil Schneiderman
- Department of Psychology, University of Miami, Coral Gables, FL, USA
| |
Collapse
|
47
|
Favre J, Vessieres E, Guihot AL, Proux C, Grimaud L, Rivron J, Garcia MC, Réthoré L, Zahreddine R, Davezac M, Fébrissy C, Adlanmerini M, Loufrani L, Procaccio V, Foidart JM, Flouriot G, Lenfant F, Fontaine C, Arnal JF, Henrion D. Membrane estrogen receptor alpha (ERα) participates in flow-mediated dilation in a ligand-independent manner. eLife 2021; 10:68695. [PMID: 34842136 PMCID: PMC8676342 DOI: 10.7554/elife.68695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) activation by estrogens prevents atheroma through its nuclear action, whereas plasma membrane-located ERα accelerates endothelial healing. The genetic deficiency of ERα was associated with a reduction in flow-mediated dilation (FMD) in one man. Here, we evaluated ex vivo the role of ERα on FMD of resistance arteries. FMD, but not agonist (acetylcholine, insulin)-mediated dilation, was reduced in male and female mice lacking ERα (Esr1-/- mice) compared to wild-type mice and was not dependent on the presence of estrogens. In C451A-ERα mice lacking membrane ERα, not in mice lacking AF2-dependent nuclear ERα actions, FMD was reduced, and restored by antioxidant treatments. Compared to wild-type mice, isolated perfused kidneys of C451A-ERα mice revealed a decreased flow-mediated nitrate production and an increased H2O2 production. Thus, endothelial membrane ERα promotes NO bioavailability through inhibition of oxidative stress and thereby participates in FMD in a ligand-independent manner.
Collapse
Affiliation(s)
- Julie Favre
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Anne-Laure Guihot
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Coralyne Proux
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Linda Grimaud
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Jordan Rivron
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Manuela Cl Garcia
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Léa Réthoré
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Rana Zahreddine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Morgane Davezac
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Chanaelle Fébrissy
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Laurent Loufrani
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Vincent Procaccio
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgium
| | - Gilles Flouriot
- INSERM U1085, IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Rennes, France
| | - Françoise Lenfant
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Coralie Fontaine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Jean-François Arnal
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Daniel Henrion
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| |
Collapse
|
48
|
Zahreddine R, Davezac M, Buscato M, Smirnova N, Laffargue M, Henrion D, Adlanmerini M, Lenfant F, Arnal JF, Fontaine C. A historical view of estrogen effect on arterial endothelial healing: From animal models to medical implication. Atherosclerosis 2021; 338:30-38. [PMID: 34785429 DOI: 10.1016/j.atherosclerosis.2021.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/18/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022]
Abstract
Endothelial barrier integrity is required for maintaining vascular homeostasis and fluid balance between the circulation and surrounding tissues. In contrast, abnormalities of endothelial cell function and loss of the integrity of the endothelial monolayer constitute a key step in the onset of atherosclerosis. Endothelial erosion is directly responsible for thrombus formation and cardiovascular events in about one-third of the cases of acute coronary syndromes. Thus, after endothelial injury, the vascular repair process is crucial to restore endothelial junctions and rehabilitate a semipermeable barrier, preventing the development of vascular diseases. Endothelial healing can be modulated by several factors. In particular, 17β-estradiol (E2), the main estrogen, improves endothelial healing, reduces neointimal accumulation of smooth muscle cells and atherosclerosis in several animal models. The aim of this review is to highlight how various experimental models enabled the progress in the cellular and molecular mechanisms underlying the accelerative E2 effect on arterial endothelial healing through the estrogen receptor (ER) α, the main receptor mediating the physiological effects of estrogens. We first summarize the different experimental procedures used to reproduce vascular injury. We then provide an overview of how the combination of transgenic mouse models impacting ERα signalling with pharmacological tools demonstrated the pivotal role of non-genomic actions of ERα in E2-induced endothelial repair. Finally, we describe recent advances in the action of selective estrogen receptor modulators (SERMs) on this beneficial vascular effect, which surprisingly involves different cell types and activates different ERα subfunctions compared to E2.
Collapse
Affiliation(s)
- Rana Zahreddine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Morgane Davezac
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Melissa Buscato
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Natalia Smirnova
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Muriel Laffargue
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Daniel Henrion
- MITOVASC Institute, CARFI Facility, INSERM U1083, UMR CNRS 6015, University of Angers, France
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1297, University of Toulouse3, Toulouse, France.
| |
Collapse
|
49
|
Baranauskas MN, Freemas JA, Tan R, Carter SJ. Moving beyond inclusion: Methodological considerations for the menstrual cycle and menopause in research evaluating effects of dietary nitrate on vascular function. Nitric Oxide 2021; 118:39-48. [PMID: 34774755 DOI: 10.1016/j.niox.2021.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/18/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
Recent reports have acknowledged the underrepresentation of women in the field of dietary nitrate (NO3-) research. Undoubtedly, greater participation from women is warranted to clarify potential sex differences in the responses to dietary NO3- interventions. However, careful consideration for the effects of sex hormones - principally 17β-estradiol - on endogenous nitric oxide (NO) synthesis and dietary NO3- reductase capacity is necessary for improved interpretation and reproducibility of such investigations. From available literature, we present a narrative review describing how hormonal variations across the menstrual cycle, as well as with menopause, may impact NO biosynthesis catalyzed by NO synthase enzymes and NO3- reduction via the enterosalivary pathway. In doing so, we address methodological considerations related to the menstrual cycle and hormonal contraceptive use relevant for the inclusion of premenopausal women along with factors to consider when testing postmenopausal women. Adherence to such methodological practices may explicate the utility of dietary NO3- supplementation as a means to improve vascular function among women across the lifespan.
Collapse
Affiliation(s)
- Marissa N Baranauskas
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA.
| | - Jessica A Freemas
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA
| | - Rachel Tan
- Department of Natural Science, Seaver College, Pepperdine University, 90263, USA
| | - Stephen J Carter
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| |
Collapse
|
50
|
Abstract
Endometriosis, characterized by macroscopic lesions in the ovaries, is a serious problem for women who desire conception. Damage to the ovarian cortex is inevitable when lesions are removed via surgery, which finally decreases the ovarian reserve, thereby accelerating the transition to the menopausal state. Soon after cessation of ovarian function, in addition to climacteric symptoms, dyslipidemia and osteopenia are known to occur in women aged >50 years. Epidemiologically, there are sex-related differences in the frequencies of dyslipidemia, hypertension, and osteoporosis. Females are more susceptible to these diseases, prevention of which is important for healthy life expectancy. Dyslipidemia and hypertension are associated with the progression of arteriosclerosis, and arteriosclerotic changes in the large and middle blood vessels are one of the main causes of myocardial and cerebral infarctions. Osteoporosis is associated with aberrant fractures in the spine and hip, which may confine the patients to the bed for long durations. Bone resorption is accelerated by activated osteoclasts, and rapid bone remodeling reduces bone mineral density. Resveratrol, a plant-derived molecule that promotes the function and expression of the sirtuin, SIRT1, has been attracting attention, and many reports have shown that resveratrol might exert cardiovascular protective effects. Preclinical reports also indicate that it can prevent bone loss and endometriosis. In this review, I have described the possible protective effects of resveratrol against arteriosclerosis, osteoporosis, and endometriosis because of its wide-ranging functions, including anti-inflammatory and antioxidative stress functions. As ovarian function inevitably declines after 40 years, intake of resveratrol can be beneficial for women with endometriosis aged <40 years.
Collapse
|