1
|
Aladelokun O, Benitez K, Wang Y, Jain A, Berardi D, Maroun G, Shen X, Roper J, Gibson J, Sumigray K, Khan SA, Johnson CH. Sex-specific effects of exogenous asparagine on colorectal tumor growth, 17β-estradiol levels, and aromatase. Pharmacol Res 2025; 215:107736. [PMID: 40228761 DOI: 10.1016/j.phrs.2025.107736] [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: 12/30/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Sex-related differences in asparagine metabolism are associated with cancer prognosis. However, the effect of exogenous asparagine on colorectal cancer (CRC) growth in men and women remains unclear. This study aims to understand the relationship between exogenous asparagine supplementation and 17β-estradiol levels and to elucidate mechanisms underlying sex-dependent signaling during CRC development. We administered asparagine intraperitoneally to tumor-bearing male and female immunodeficient Rag2/Il2RG (R2G2) mice. Asparagine supplementation caused a significant increase in tumor asparagine levels in both the tumor-bearing male and female R2G2 mice but increased serum estradiol levels and suppressed tumor growth in female R2G2 mice only. Additionally, we combined transcriptome, metabolome, and immunochemical analyses, and found that intraperitoneal asparagine treatment induced sex-dependent intra-tumoral metabolic changes to asparagine, aspartate, glutamine and glutamate levels. We observed that in females, exogenous asparagine exerts a negative feed-back effect on de novo asparagine synthesis and is associated with the activation of a sub-population of macrophages that may secrete 17β-estradiol via an aromatase or cytochrome P450 family 19 (CYP19)-dependent mechanism. Conversely, in male mice, asparagine treatment augments tumor growth, and is related to decreased numbers of macrophages, and a reduction in CYP19-mediated 17β-estradiol secretion . Overall, our results reveal a novel and sex-specific role for exogenous asparagine during cancer progression and underscores the importance of understanding mechanisms that control asparagine biosynthesis.
Collapse
Affiliation(s)
- Oladimeji Aladelokun
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Katherine Benitez
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yuying Wang
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Abhishek Jain
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Domenica Berardi
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Georgio Maroun
- Department of Molecular Biochemistry and Biophysics, Yale University, New Haven, CT, USA
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, NC, USA
| | - Joanna Gibson
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sajid A Khan
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
2
|
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
|
3
|
Wang Z, Liu J, Chen Y, Tang Y, Chen T, Zhou C, Wang S, Chang R, Chen Z, Yang W, Guo Z, Chen T. From physiology to pathology: Emerging roles of GPER in cardiovascular disease. Pharmacol Ther 2025; 267:108801. [PMID: 39889969 DOI: 10.1016/j.pharmthera.2025.108801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Cardiovascular diseases (CVDs) are among the leading causes of death globally and pose a significant threat to public health. Factors such as prolonged high cholesterol levels, diabetes, smoking, unhealthy diet, and genetic predisposition could contribute to the occurrence and development of CVDs. Common CVDs include hypertension (HTN), atherosclerosis (AS), myocardial infarction (MI), myocardial ischemia-reperfusion injury (MIRI), heart failure (HF) and arrhythmia. Estrogen is recognized for its cardiovascular protective effects, resulting in lower incidence and mortality rates of CVDs in premenopausal women compared to men. The G protein-coupled estrogen receptor (GPER), a G protein-coupled receptor with a seven-transmembrane structure, exhibits unique structural characteristics and widespread tissue distribution. GPER activates intracellular signaling pathways through its interaction with G proteins, mediating estrogen's biological effects and participating in the regulation of cardiovascular function, metabolic balance, and nervous system. Although recent research has highlighted the significant role of GPER in the cardiovascular system, its specific mechanisms remain unclear. Therefore, this review summarizes the latest research on GPER in CVDs, including its fundamental characteristics, physiological functions in the cardiovascular system, and its roles and potential therapeutic applications in common CVDs such as HTN, AS, MI, MIRI, HF and arrhythmia. Exploring GPER's positive effects on cardiovascular health will provide new strategies and research directions for the treatment of CVDs.
Collapse
Affiliation(s)
- Zixuan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junren Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ying Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yi Tang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ting Chen
- Hunan University of Chinese Medicine, The College of Acupuncture & Moxibustion and Tuina, Changsha 410208, China
| | - Chang Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shuo Wang
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617.China
| | - Ranbo Chang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhongshuai Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Wenqing Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhen Guo
- Hunan Provincial Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha 410219, China
| | - Ting Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China; Hunan Provincial Key Laboratory of Traditional Chinese Medicine Powder and Innovative drug Research, Changsha 410208, China.
| |
Collapse
|
4
|
Kamau AN, Sakamuri A, Okoye DO, Sengottaian D, Cannon J, Guerrero-Millan J, Sullivan JC, Miller KS, Liu Y, Ogola BO. Sexual dimorphism in the downregulation of extracellular matrix genes contributes to aortic stiffness in female mice. Am J Physiol Heart Circ Physiol 2025; 328:H472-H483. [PMID: 39873616 DOI: 10.1152/ajpheart.00432.2024] [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: 06/28/2024] [Revised: 07/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
The contribution of sex hormones to cardiovascular disease, including arterial stiffness, is established; however, the role of sex chromosome interaction with sex hormones, particularly in women, is lagging. Arterial stiffness depends on the intrinsic properties and transmural wall geometry that comprise a network of cells and extracellular matrix (ECM) proteins expressed in a sex-dependent manner. In this study, we used four-core genotype (FCG) mice to determine the relative contribution of sex hormones versus sex chromosomes or their interaction with arterial stiffness. Gonadal intact FCG mice included females (F) and males (M) with either XX or XY sex chromosomes (n = 9-11/group). We isolated the thoracic aorta, and a tissue puller was used to assess structural resistance to changes in shape under control, collagenase, or elastase conditions. We determined histological collagen area fraction and evaluated aortic ECM genes by PCR microarrays followed by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Stress-strain curves showed higher elastic modulus (P < 0.001), denoting decreased distensibility in XXF compared with XYF aortas, which were significantly reversed by collagenase and elastase treatments (P < 0.01). Aortic gene expression analysis indicated a significant reduction in Emilin1, Thbs2, and Icam1 in the XXF versus XYF aorta (P < 0.05). Uniaxial stretching of XXF aortic vascular smooth muscle cells indicated decreased Thbs2, Ctnna1, and Ecm1 genes. We observed a significant (P < 0.05) reduction in Masson's trichrome staining in collagenase but not elastase-treated aortic rings compared with the control. The increased aortic elastic modulus in XXF compared with XYF mice suggests a decrease in aortic distensibility mediated by a reduction in ECM genes.NEW & NOTEWORTHY FCG mice model can segregate vascular phenotypes by sex hormones, sex chromosomes, and their interaction. We show increased aortic stiffening in XX versus XY female mice and decreased ECM genes, suggesting decreased distensibility and impaired mechanotransduction in XXF versus XYF mice aortas. Therefore, the XX versus XY differences imply a unique role for sex chromosomes in regulating aortic integrity and ECM genes in female mice.
Collapse
Affiliation(s)
- Anne N Kamau
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Anil Sakamuri
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Delphine O Okoye
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Divya Sengottaian
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jennifer Cannon
- Department of Biology, Augusta University, Augusta, Georgia, United States
| | - Josefa Guerrero-Millan
- Department of Physics and Biophysics, Augusta University, Augusta, Georgia, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Kristin S Miller
- Department of Bioengineering, The University of Texas at Dallas, Richardson, Texas, United States
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, Texas, United States
- Department of Obstetrics & Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Benard O Ogola
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
5
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
6
|
Wendt TS, Ansar S, Gonzales RJ. OxLDL/LOX-1 mediated sex, age, stiffness, and endothelial dependent alterations in mouse thoracic aortic vascular reactivity. Front Physiol 2024; 15:1471272. [PMID: 39563936 PMCID: PMC11573510 DOI: 10.3389/fphys.2024.1471272] [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: 07/27/2024] [Accepted: 10/03/2024] [Indexed: 11/21/2024] Open
Abstract
Elevated plasma levels of oxidized low-density lipoprotein (oxLDL) are a risk factor and key component that accelerates and worsens cardiovascular disease fueling inflammation, plaque buildup and vascular damage. OxLDL can elicit its detrimental action via lectin-like oxLDL receptor 1 (LOX-1). In this study, we determined whether oxLDL, via LOX-1, alters aortic vascular reactivity and determined if sex and age differences exist. Thoracic aortic endothelium-intact or -denuded ring segments were isolated from 7 to 12 months old intact C57BL/6J female and male mice and pre-incubated with oxLDL ex vivo (50ug/dL; 2 h). Using wire myography, cumulative concentration-response curves to phenylephrine (PE) were generated to determine contractile responses. From these curves, the EC50 was determined and used to contract rings to assess acetylcholine (ACh) dependent relaxation. Calculated aortic stiffness and remodeling were also assessed. BI-0115 (10 μ M; selective LOX-1 inhibitor) was used to determine LOX-1 dependence. We observed differential sex, age, endothelial cell, and LOX-1 dependent alterations to the efficacy of PE-induced contractile responses and ACh-mediated vasorelaxation in thoracic aortic rings following oxLDL exposure. Additionally, we observed a distinct sex and age effect on thoracic aortic stiffness following exposure to oxLDL. There was also a sex effect on calculated vessel diameter, as well as an age effect on oxLDL-mediated aortic remodeling that was LOX-1 dependent. Thus, LOX-1 inhibition and the resulting attenuation of oxLDL/endothelial-mediated alterations in aortic function suggests that there are differential sex differences in the role of oxLDL/LOX-1 in the thoracic aorta of middle-aged male and female mice. NEW and NOTEWORTHY. We investigated the effects of oxLDL via the LOX-1 receptor on murine thoracic aortic vasoreactivity, stiffness, and remodeling across age and sex. Acute exposure to oxLDL led to altered vasoreactivity, endothelial dysfunction, and changes in aortic stiffness and remodeling. These effects were in-part age, sex, endothelial, and LOX-1 dependent. This study reveals potential complex interactions in oxLDL/LOX-1-mediated vascular responses that could serve as potential therapeutic intervention for vascular diseases such as atherosclerosis and stroke.
Collapse
Affiliation(s)
- Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona, Phoenix, AZ, United States
| | - Saema Ansar
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona, Phoenix, AZ, United States
| |
Collapse
|
7
|
Guldan M, Unlu S, Abdel-Rahman SM, Ozbek L, Gaipov A, Covic A, Soler MJ, Covic A, Kanbay M. Understanding the Role of Sex Hormones in Cardiovascular Kidney Metabolic Syndrome: Toward Personalized Therapeutic Approaches. J Clin Med 2024; 13:4354. [PMID: 39124622 PMCID: PMC11312746 DOI: 10.3390/jcm13154354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiovascular kidney metabolic (CKM) syndrome represents a complex interplay of cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic comorbidities, posing a significant public health challenge. Gender exerts a critical influence on CKM syndrome, affecting the disease severity and onset through intricate interactions involving sex hormones and key physiological pathways such as the renin-angiotensin system, oxidative stress, inflammation, vascular disease and insulin resistance. It is widely known that beyond the contribution of traditional risk factors, men and women exhibit significant differences in CKM syndrome and its components, with distinct patterns observed in premenopausal women and postmenopausal women compared to men. Despite women generally experiencing a lower incidence of CVD, their outcomes following cardiovascular events are often worse compared to men. The disparities also extend to the treatment approaches for kidney failure, with a higher prevalence of dialysis among men despite women exhibiting higher rates of CKD. The impact of endogenous sex hormones, the correlations between CKM and its components, as well as the long-term effects of treatment modalities using sex hormones, including hormone replacement therapies and gender-affirming therapies, have drawn attention to this topic. Current research on CKM syndrome is hindered by the scarcity of large-scale studies and insufficient integration of gender-specific considerations into treatment strategies. The underlying mechanisms driving the gender disparities in the pathogenesis of CKM syndrome, including the roles of estrogen, progesterone and testosterone derivatives, remain poorly understood, thus limiting their application in personalized therapeutic interventions. This review synthesizes existing knowledge to clarify the intricate relationship between sex hormones, gender disparities, and the progression of CVD within CKM syndrome. By addressing these knowledge gaps, this study aims to guide future research efforts and promote tailored approaches for effectively managing CKD syndrome.
Collapse
Affiliation(s)
- Mustafa Guldan
- Department of Medicine, Koç University School of Medicine, 34450 Istanbul, Turkey; (M.G.); (S.U.); (S.M.A.-R.); (L.O.)
| | - Selen Unlu
- Department of Medicine, Koç University School of Medicine, 34450 Istanbul, Turkey; (M.G.); (S.U.); (S.M.A.-R.); (L.O.)
| | - Sama Mahmoud Abdel-Rahman
- Department of Medicine, Koç University School of Medicine, 34450 Istanbul, Turkey; (M.G.); (S.U.); (S.M.A.-R.); (L.O.)
| | - Laşin Ozbek
- Department of Medicine, Koç University School of Medicine, 34450 Istanbul, Turkey; (M.G.); (S.U.); (S.M.A.-R.); (L.O.)
| | - Abduzhappar Gaipov
- Department of Medicine, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Andreea Covic
- Department of Nephrology, Grigore T. Popa University of Medicine, 700115 Iasi, Romania;
| | - Maria José Soler
- Nephrology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Research, 08035 Barcelona, Spain;
- Centro de Referencia en Enfermedad, Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), RICORS2040 (Kidney Disease), 08003 Barcelona, Spain
- GEENDIAB (Grupo Español de Estudio de la Nefropatía Diabética), 39008 Santander, Spain
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa University of Medicine, 700115 Iasi, Romania;
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koç University School of Medicine, 34450 Istanbul, Turkey;
| |
Collapse
|
8
|
Aladelokun O, Lu L, Zheng J, Yan H, Jain A, Gibson J, Khan SA, Johnson CH. Growth characteristics of HCT116 xenografts lacking asparagine synthetase vary according to sex. Hum Genomics 2024; 18:67. [PMID: 38886847 PMCID: PMC11184737 DOI: 10.1186/s40246-024-00635-3] [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: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Sex-related differences in colorectal (CRC) incidence and mortality are well-documented. However, the impact of sex on metabolic pathways that drive cancer growth is not well understood. High expression of asparagine synthetase (ASNS) is associated with inferior survival for female CRC patients only. Here, we used a CRISPR/Cas9 technology to generate HCT116 ASNS-/- and HCT 116 ASNS+/+ cancer cell lines. We examine the effects of ASNS deletion on tumor growth and the subsequent rewiring of metabolic pathways in male and female Rag2/IL2RG mice. RESULTS ASNS loss reduces cancer burden in male and female tumor-bearing mice (40% reduction, q < 0.05), triggers metabolic reprogramming including gluconeogenesis, but confers a survival improvement (30 days median survival, q < 0.05) in female tumor-bearing mice alone. Transcriptomic analyses revealed upregulation of G-protein coupled estrogen receptor (GPER1) in tumors from male and female mice with HCT116 ASNS-/- xenograft. Estradiol activates GPER1 in vitro in the presence of ASNS and suppresses tumor growth. CONCLUSIONS Our study indicates that inferior survival for female CRC patients with high ASNS may be due to metabolic reprogramming that sustains tumor growth. These findings have translational relevance as ASNS/GPER1 signaling could be a future therapeutic target to improve the survival of female CRC patients.
Collapse
Affiliation(s)
- Oladimeji Aladelokun
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Jie Zheng
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Abhishek Jain
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Joanna Gibson
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Sajid A Khan
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
9
|
Sakamuri A, Visniauskas B, Kilanowski-Doroh I, McNally AB, Imulinde A, Kamau A, Sengottaian D, McLachlan J, Anguera M, Mauvais-Jarvis F, Lindsey SH, Ogola BO. Testosterone deficiency promotes arterial stiffening independent of sex chromosome complement. Biol Sex Differ 2024; 15:46. [PMID: 38845040 PMCID: PMC11155160 DOI: 10.1186/s13293-024-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Sex hormones and sex chromosomes play a vital role in cardiovascular disease. Testosterone plays a crucial role in men's health. Lower testosterone level is associated with cardiovascular and cardiometabolic diseases, including inflammation, atherosclerosis, and type 2 diabetes. Testosterone replacement is beneficial or neutral to men's cardiovascular health. Testosterone deficiency is associated with cardiovascular events. Testosterone supplementation to hypogonadal men improves libido, increases muscle strength, and enhances mood. We hypothesized that sex chromosomes (XX and XY) interaction with testosterone plays a role in arterial stiffening. METHODS We used four core genotype male mice to understand the inherent contribution of sex hormones and sex chromosome complement in arterial stiffening. Age-matched mice were either gonadal intact or castrated at eight weeks plus an additional eight weeks to clear endogenous sex hormones. This was followed by assessing blood pressure, pulse wave velocity, echocardiography, and ex vivo passive vascular mechanics. RESULTS Arterial stiffening but not blood pressure was more significant in castrated than testes-intact mice independent of sex chromosome complement. Castrated mice showed a leftward shift in stress-strain curves and carotid wall thinning. Sex chromosome complement (XX) in the absence of testosterone increased collagen deposition in the aorta and Kdm6a gene expression. CONCLUSION Testosterone deprivation increases arterial stiffening and vascular wall remodeling. Castration increases Col1α1 in male mice with XX sex chromosome complement. Our study shows decreased aortic contractile genes in castrated mice with XX than XY sex chromosomes.
Collapse
Affiliation(s)
- Anil Sakamuri
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | | | | | - Ariane Imulinde
- Department of Pharmacology, Tulane University, New Orleans, LA, USA
| | - Anne Kamau
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Divya Sengottaian
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - John McLachlan
- Department of Pharmacology, Tulane University, New Orleans, LA, USA
| | - Montserrat Anguera
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Franck Mauvais-Jarvis
- Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA
- Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA, USA
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, USA
- Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA
| | - Benard O Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
10
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Sugi AI, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs From Vascular Aging and Is Reversed by GPER Activation. Hypertension 2024; 81:e51-e62. [PMID: 38445498 PMCID: PMC11023783 DOI: 10.1161/hypertensionaha.123.22024] [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: 09/07/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging and whether activation of the G-protein-coupled estrogen receptor could reverse stiffness. METHODS Female C57Bl/6J mice were ovariectomized at 10 weeks of age or aged to 52 weeks, and some mice were treated with G-protein-coupled estrogen receptor agonists. RESULTS Ovariectomy and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while ovariectomy increased material stiffness without altering vascular geometry. RNA-sequencing analysis revealed that ovariectomy downregulated smooth muscle contractile genes. The enantiomerically pure G-protein-coupled estrogen receptor agonist, LNS8801, reversed stiffness in ovariectomy mice to a greater degree than the racemic agonist G-1. In summary, ovariectomy and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling, while ovariectomy-induced material stiffness independent of geometry and a loss of the contractile phenotype. CONCLUSIONS This study enhances our understanding of the impact of estrogen loss on vascular health in a murine model and warrants further studies to examine the ability of LNS8801 to improve vascular health in menopausal women.
Collapse
Affiliation(s)
| | | | - Tristen Wong
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Bruna Visniauskas
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | | | - Chase Richard
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Zaidmara Diaz
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Alec Horton
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | - Benard O. Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
11
|
Sullivan BP, Collins BC, McMillin SL, Toussaint E, Stein CZ, Spangenburg EE, Lowe DA. Ablation of skeletal muscle estrogen receptor alpha impairs contractility in male mice. J Appl Physiol (1985) 2024; 136:764-773. [PMID: 38328824 PMCID: PMC11286273 DOI: 10.1152/japplphysiol.00714.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Estradiol and estrogen receptor α (ERα) have been shown to be important for the maintenance of skeletal muscle strength in females; however, little is known about the roles of estradiol and ERα in male muscle. The purpose of this study was to determine if skeletal muscle ERα is required for optimal contractility in male mice. We hypothesize that reduced ERα in skeletal muscle impairs contractility in male mice. Skeletal muscle-specific knockout (skmERαKO) male mice exhibited reduced strength across multiple muscles and several contractile parameters related to force generation and kinetics compared with wild-type littermates (skmERαWT). Isolated EDL muscle-specific isometric tetanic force, peak twitch force, peak concentric and peak eccentric forces, as well as the maximal rates of force development and relaxation were 11%-21% lower in skmERαKO compared with skmERαWT mice. In contrast, isolated soleus muscles from skmERαKO mice were not affected. In vivo peak torque of the anterior crural muscles was 20% lower in skmERαKO compared with skmERαWT mice. Muscle masses, contractile protein contents, fiber types, phosphorylation of the myosin regulatory light chain, and caffeine-elicited force did not differ between muscles of skmERαKO and skmERαWT mice, suggesting that strength deficits were not due to size, composition, or calcium release components of muscle contraction. These results indicate that in male mice, reduced skeletal muscle ERα blunts contractility to a magnitude similar to that previously reported in females; however, the mechanism may be sexually dimorphic.NEW & NOTEWORTHY We comprehensively measured in vitro and in vivo contractility of leg muscles with reduced estrogen receptor α (ERα) in male mice and reported that force generation and contraction kinetics are impaired. In contrast to findings in females, phosphorylation of myosin regulatory light chain cannot account for low force production in male skeletal muscle ERα knockout mice. These results indicate that ERα is required for optimal contractility in males and females but via sexually dimorphic means.
Collapse
Affiliation(s)
- Brian P Sullivan
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Brittany C Collins
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Shawna L McMillin
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Elise Toussaint
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Clara Z Stein
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina, United States
| | - Dawn A Lowe
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
12
|
Reeve EH, Barnes JN, Moir ME, Walker AE. Impact of arterial stiffness on cerebrovascular function: a review of evidence from humans and preclincal models. Am J Physiol Heart Circ Physiol 2024; 326:H689-H704. [PMID: 38214904 PMCID: PMC11221809 DOI: 10.1152/ajpheart.00592.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
With advancing age, the cerebral vasculature becomes dysfunctional, and this dysfunction is associated with cognitive decline. However, the initiating cause of these age-related cerebrovascular impairments remains incompletely understood. A characteristic feature of the aging vasculature is the increase in stiffness of the large elastic arteries. This increase in arterial stiffness is associated with elevated pulse pressure and blood flow pulsatility in the cerebral vasculature. Evidence from both humans and rodents supports that increases in large elastic artery stiffness are associated with cerebrovascular impairments. These impacts on cerebrovascular function are wide-ranging and include reductions in global and regional cerebral blood flow, cerebral small vessel disease, endothelial cell dysfunction, and impaired perivascular clearance. Furthermore, recent findings suggest that the relationship between arterial stiffness and cerebrovascular function may be influenced by genetics, specifically APOE and NOTCH genotypes. Given the strength of the evidence that age-related increases in arterial stiffness have deleterious impacts on the brain, interventions that target arterial stiffness are needed. The purpose of this review is to summarize the evidence from human and rodent studies, supporting the role of increased arterial stiffness in age-related cerebrovascular impairments.
Collapse
Affiliation(s)
- Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jill N Barnes
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - M Erin Moir
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
13
|
Lindsey ML, Usselman CW, Ripplinger CM, Carter JR, DeLeon-Pennell KY. Sex as a biological variable for cardiovascular physiology. Am J Physiol Heart Circ Physiol 2024; 326:H459-H469. [PMID: 38099847 PMCID: PMC11219053 DOI: 10.1152/ajpheart.00727.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 02/03/2024]
Abstract
There have been ongoing efforts by federal agencies and scientific communities since the early 1990s to incorporate sex and/or gender in all aspects of cardiovascular research. Scientific journals provide a critical function as change agents to influence transformation by encouraging submissions for topic areas, and by setting standards and expectations for articles submitted to the journal. As part of ongoing efforts to advance sex and gender in cardiovascular physiology research, the American Journal of Physiology-Heart and Circulatory Physiology recently launched a call for papers on Considering Sex as a Biological Variable. This call was an overwhelming success, resulting in 78 articles published in this collection. This review summarizes the major themes of the collection, including Sex as a Biological Variable Within: Endothelial Cell and Vascular Physiology, Cardiovascular Immunity and Inflammation, Metabolism and Mitochondrial Energy, Extracellular Matrix Turnover and Fibrosis, Neurohormonal Signaling, and Cardiovascular Clinical and Epidemiology Assessments. Several articles also focused on establishing rigor and reproducibility of key physiological measurements involved in cardiovascular health and disease, as well as recommendations and considerations for study design. Combined, these articles summarize our current understanding of sex and gender influences on cardiovascular physiology and pathophysiology and provide insight into future directions needed to further expand our knowledge.
Collapse
Affiliation(s)
- Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Charlotte W Usselman
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Crystal M Ripplinger
- Department of Pharmacology, UC Davis School of Medicine, Davis, California, United States
| | - Jason R Carter
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, United States
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, School of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| |
Collapse
|
14
|
Horton AC, Wilkinson MM, Kilanowski-Doroh I, Dong Z, Liu J, Ogola BO, Visniauskas B, Lindsey SH. Dihydrotestosterone induces arterial stiffening in female mice. Biol Sex Differ 2024; 15:9. [PMID: 38263051 PMCID: PMC10804721 DOI: 10.1186/s13293-024-00586-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Androgens are important sex hormones in both men and women and are supplemented when endogenous levels are low, for gender transitioning, or to increase libido. Androgens also circulate at higher levels in women with polycystic ovarian syndrome, a condition that increases the risk for cardiovascular diseases including hypertension and arterial stiffness. Since our previous work shows an important role for the G protein-coupled estrogen receptor (GPER) in arterial stiffness, we hypothesized that other hormones including androgens may impact arterial stiffness in female mice via downregulation of GPER. METHODS The impact of the non-aromatizable androgen dihydrotestosterone (DHT), the glucocorticoid dexamethasone, and the progestin medroxyprogesterone acetate (all 100 nM for 24 h) on GPER and ERα expression was assessed in cultured vascular smooth muscle cells using droplet digital PCR (ddPCR). To assess the in vivo impact of the DHT-induced downregulation of GPER, female ovary-intact C57Bl/6 mice at 15-16 weeks of age were treated with silastic capsules containing DHT for 4 weeks, one with a dosage expected to mimic human male DHT levels and another to double the expected human concentration (n = 8-9/group). RESULTS In cultured vascular smooth muscle cells, GPER mRNA was decreased by DHT (P = 0.001) but was not impacted by dexamethasone or medroxyprogesterone. In contrast, ERα expression in cultured cells was significantly suppressed by all three hormones (P < 0.0001). In control mice or mice treated with a single or double dose of DHT, a dose-dependent increase in body weight was observed (control 22 ± 2 g, single dose 24 ± 2 g, double dose 26 ± 2 g; P = 0.0002). Intracarotid stiffness measured via pulse wave velocity showed a more than two-fold increase in both DHT-treated groups (control 1.9 ± 0.3 m/s, single dose 4.3 ± 0.8 m/s, double dose 4.8 ± 1.0 m/s). This increase in arterial stiffness occurred independent of changes in blood pressure (P = 0.59). Histological analysis of aortic sections using Masson's trichrome showed a significant decrease in collagen between the control group (24 ± 5%) and the double dose group (17 ± 3%, P = 0.007), despite no changes in aortic wall thickness or smooth muscle content. Lastly, ddPCR showed that in vivo DHT treatment decreased aortic expression of both GPER (control 20 ± 5, single dose 10.5 ± 5.6, double dose 10 ± 4 copies/ng; P = 0.001) and ERα (control 54 ± 2, single dose 24 ± 13, and double dose 23 ± 12 copies/ng; P = 0.003). CONCLUSIONS These findings indicate that androgen promotes arterial stiffening and cardiovascular damage in female mice and is associated with decreased estrogen receptor expression. These data are important for transgender men, women using testosterone for fitness or reduced libido, as well as patients with polycystic ovarian syndrome.
Collapse
Affiliation(s)
- Alec C Horton
- Department of Pharmacology and Tulane Brain Institute, Tulane School of Medicine, New Orleans, LA, USA
| | - Mary M Wilkinson
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA, USA
| | | | - Zhejun Dong
- Department of Pediatrics, Tulane School of Medicine, Hayward Genetics Center, New Orleans, LA, USA
| | - Jiao Liu
- Department of Pediatrics, Tulane School of Medicine, Hayward Genetics Center, New Orleans, LA, USA
| | - Benard O Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Bruna Visniauskas
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology and Tulane Brain Institute, Tulane School of Medicine, New Orleans, LA, USA.
- Tulane Center of Excellence in Sex-Based Biology and Medicine, New Orleans, LA, USA.
| |
Collapse
|
15
|
Usselman CW, Lindsey ML, Robinson AT, Habecker BA, Taylor CE, Merryman WD, Kimmerly D, Bender JR, Regensteiner JG, Moreau KL, Pilote L, Wenner MM, O'Brien M, Yarovinsky TO, Stachenfeld NS, Charkoudian N, Denfeld QE, Moreira-Bouchard JD, Pyle WG, DeLeon-Pennell KY. Guidelines on the use of sex and gender in cardiovascular research. Am J Physiol Heart Circ Physiol 2024; 326:H238-H255. [PMID: 37999647 PMCID: PMC11219057 DOI: 10.1152/ajpheart.00535.2023] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
In cardiovascular research, sex and gender have not typically been considered in research design and reporting until recently. This has resulted in clinical research findings from which not only all women, but also gender-diverse individuals have been excluded. The resulting dearth of data has led to a lack of sex- and gender-specific clinical guidelines and raises serious questions about evidence-based care. Basic research has also excluded considerations of sex. Including sex and/or gender as research variables not only has the potential to improve the health of society overall now, but it also provides a foundation of knowledge on which to build future advances. The goal of this guidelines article is to provide advice on best practices to include sex and gender considerations in study design, as well as data collection, analysis, and interpretation to optimally establish rigor and reproducibility needed to inform clinical decision-making and improve outcomes. In cardiovascular physiology, incorporating sex and gender is a necessary component when optimally designing and executing research plans. The guidelines serve as the first guidance on how to include sex and gender in cardiovascular research. We provide here a beginning path toward achieving this goal and improve the ability of the research community to interpret results through a sex and gender lens to enable comparison across studies and laboratories, resulting in better health for all.
Collapse
Affiliation(s)
- Charlotte W Usselman
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Austin T Robinson
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Chloe E Taylor
- School of Health Sciences, Western Sydney University, Sydney, New South Wales, Australia
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Derek Kimmerly
- Autonomic Cardiovascular Control and Exercise Laboratory, Division of Kinesiology, School of Health and Human Performance, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey R Bender
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Judith G Regensteiner
- Divisions of General Internal Medicine and Cardiology, Department of Medicine, Ludeman Family Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kerrie L Moreau
- Division of Geriatrics, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Eastern Colorado Health Care System, Geriatric Research Education and Clinical Center, Aurora, Colorado, United States
| | - Louise Pilote
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| | - Myles O'Brien
- School of Physiotherapy and Department of Medicine, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Timur O Yarovinsky
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Nina S Stachenfeld
- John B. Pierce Laboratory, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Nisha Charkoudian
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Quin E Denfeld
- School of Nursing and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Jesse D Moreira-Bouchard
- Q.U.E.E.R. Lab, Programs in Human Physiology, Department of Health Sciences, Boston University College of Health and Rehabilitation Sciences: Sargent College, Boston, Massachusetts, United States
| | - W Glen Pyle
- IMPART Team Canada Network, Dalhousie Medicine, Saint John, New Brunswick, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kristine Y DeLeon-Pennell
- School of Medicine, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| |
Collapse
|
16
|
Behringer EJ. Impact of aging on vascular ion channels: perspectives and knowledge gaps across major organ systems. Am J Physiol Heart Circ Physiol 2023; 325:H1012-H1038. [PMID: 37624095 PMCID: PMC10908410 DOI: 10.1152/ajpheart.00288.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Individuals aged ≥65 yr will comprise ∼20% of the global population by 2030. Cardiovascular disease remains the leading cause of death in the world with age-related endothelial "dysfunction" as a key risk factor. As an organ in and of itself, vascular endothelium courses throughout the mammalian body to coordinate blood flow to all other organs and tissues (e.g., brain, heart, lung, skeletal muscle, gut, kidney, skin) in accord with metabolic demand. In turn, emerging evidence demonstrates that vascular aging and its comorbidities (e.g., neurodegeneration, diabetes, hypertension, kidney disease, heart failure, and cancer) are "channelopathies" in large part. With an emphasis on distinct functional traits and common arrangements across major organs systems, the present literature review encompasses regulation of vascular ion channels that underlie blood flow control throughout the body. The regulation of myoendothelial coupling and local versus conducted signaling are discussed with new perspectives for aging and the development of chronic diseases. Although equipped with an awareness of knowledge gaps in the vascular aging field, a section has been included to encompass general feasibility, role of biological sex, and additional conceptual and experimental considerations (e.g., cell regression and proliferation, gene profile analyses). The ultimate goal is for the reader to see and understand major points of deterioration in vascular function while gaining the ability to think of potential mechanistic and therapeutic strategies to sustain organ perfusion and whole body health with aging.
Collapse
Affiliation(s)
- Erik J Behringer
- Basic Sciences, Loma Linda University, Loma Linda, California, United States
| |
Collapse
|
17
|
Sakamuri A, Visniauskas B, Kilanowski-Doroh I, McNally A, Imulinde-Sugi A, Kamau A, Sengottaian D, McLachlan J, Anguera M, Mauvais-Jarvis F, Lindsey S, Ogola BO. Testosterone Deficiency Promotes Arterial Stiffening Independent of Sex Chromosome Complement. RESEARCH SQUARE 2023:rs.3.rs-3370040. [PMID: 37886462 PMCID: PMC10602149 DOI: 10.21203/rs.3.rs-3370040/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Background Testosterone plays a vital role in men's health. Lower testosterone level is associated with cardiovascular and cardiometabolic diseases, including inflammation, atherosclerosis, and type 2 diabetes. Testosterone replacement is beneficial or neutral to men's cardiovascular health. Testosterone deficiency is associated with cardiovascular events. Testosterone supplementation to hypogonadal men improves libido, increases muscle strength, and enhances mood. We hypothesized that sex chromosomes (XX and XY) interaction with testosterone plays a role in arterial stiffening. Methods We used four core genotype male mice to understand the inherent contribution of sex hormones and sex chromosome complement in arterial stiffening. Age-matched mice were either gonadal intact or castrated for eight weeks, followed by an assessment of blood pressure, pulse wave velocity, echocardiography, and ex vivo passive vascular mechanics. Results Arterial stiffening but not blood pressure was more significant in castrated than testes-intact mice independent of sex chromosome complement. Castrated mice showed a leftward shift in stress-strain curves and carotid wall thinning. Sex chromosome complement (XX) in the absence of testosterone increased collagen deposition in the aorta and Kdm6a gene expression. Conclusion Testosterone deprivation increases arterial stiffening and vascular wall remodeling. Castration increases Col1α1 in male mice with XX sex chromosome complement. Our study shows decreased aortic contractile genes in castrated mice with XX than XY sex chromosomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Anne Kamau
- Augusta University Medical College of Georgia
| | | | | | | | | | | | | |
Collapse
|
18
|
Holly D, Kim H, Woodman CR, Massett MP. Genetic background influences arterial vasomotor function in male and female mice. Physiol Rep 2023; 11:e15824. [PMID: 37771071 PMCID: PMC10539628 DOI: 10.14814/phy2.15824] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 09/30/2023] Open
Abstract
The purpose of this study was to assess the influence of genetic background and sex on nitric oxide (NO)-mediated vasomotor function in arteries from different vascular territories. Vasomotor function was assessed in thoracic aorta, abdominal aorta, carotid arteries, and femoral arteries from the following mouse strains: SJL/J, DBA/2J, NZW/LacJ, and C57BL/6J. Contractile responses were assessed using the α1-adrenergic receptor agonist phenylephrine (PE, 10-9 -10-5 M). Vasorelaxation responses were assessed by examining relaxation to an endothelium-dependent vasodilator acetylcholine (ACh, 10-9 -10-5 M) and an endothelium-independent vasodilator sodium nitroprusside (SNP, 10-9 -10-5 M). To evaluate the role of NO, relaxation responses to ACh and SNP were assessed in the absence or presence of a nitric oxide synthase inhibitor (N omega-nitro-l-arginine methyl ester hydrochloride: 10-4 M). Vasomotor responses to ACh and PE varied across strains and among the arteries tested with some strains exhibiting artery-specific impairment. Results indicated some concentration-response heterogeneity in response to ACh and SNP between vessels from females and males, but no significant differences in responses to PE. Collectively, these findings indicate that vasomotor responses vary by genetic background, sex, and artery type.
Collapse
Affiliation(s)
- Dylan Holly
- Department of Kinesiology and Sport ManagementTexas A&M UniversityCollege StationTexasUSA
| | - Hyoseon Kim
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| | - Christopher R. Woodman
- Department of Kinesiology and Sport ManagementTexas A&M UniversityCollege StationTexasUSA
| | - Michael P. Massett
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| |
Collapse
|
19
|
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
|
20
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Imulinde Sugi A, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs from Vascular Aging and is Reversed by GPER Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552881. [PMID: 37645992 PMCID: PMC10462036 DOI: 10.1101/2023.08.10.552881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging, and whether activation of the G protein-coupled estrogen receptor (GPER) could reverse stiffness. Female C57Bl/6J mice were ovariectomized (OVX) at 10 weeks of age or aged to 52 weeks, and some mice were treated with GPER agonists. OVX and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while OVX increased material stiffness without altering vascular geometry. RNA-Seq analysis revealed that OVX downregulated smooth muscle contractile genes. The enantiomerically pure GPER agonist, LNS8801, reversed stiffness in OVX mice to a greater degree than the racemic agonist G-1. In summary, OVX and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling while OVX induced material stiffness independent of geometry and a loss of the contractile phenotype. This study helps to further our understanding of the impact of menopause on vascular health and identifies LNS8801 as a potential therapy to counteract this detrimental process in women.
Collapse
|
21
|
Visniauskas B, Kilanowski-Doroh I, Ogola BO, Mcnally AB, Horton AC, Imulinde Sugi A, Lindsey SH. Estrogen-mediated mechanisms in hypertension and other cardiovascular diseases. J Hum Hypertens 2023; 37:609-618. [PMID: 36319856 PMCID: PMC10919324 DOI: 10.1038/s41371-022-00771-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally for men and women. Premenopausal women have a lower incidence of hypertension and other cardiovascular events than men of the same age, but diminished sex differences after menopause implicates 17-beta-estradiol (E2) as a protective agent. The cardioprotective effects of E2 are mediated by nuclear estrogen receptors (ERα and ERβ) and a G protein-coupled estrogen receptor (GPER). This review summarizes both established as well as emerging estrogen-mediated mechanisms that underlie sex differences in the vasculature during hypertension and CVD. In addition, remaining knowledge gaps inherent in the association of sex differences and E2 are identified, which may guide future clinical trials and experimental studies in this field.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ariane Imulinde Sugi
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center of Excellence in Sex-Based Biology and Medicine, New Orleans, LA, USA.
- Tulane Brain Institute, New Orleans, LA, USA.
| |
Collapse
|
22
|
Visniauskas B, Reverte V, Abshire CM, Ogola BO, Rosales CB, Galeas-Pena M, Sure VN, Sakamuri SSVP, Harris NR, Kilanowski-Doroh I, Mcnally AB, Horton AC, Zimmerman M, Katakam PVG, Lindsey SH, Prieto MC. High-plasma soluble prorenin receptor is associated with vascular damage in male, but not female, mice fed a high-fat diet. Am J Physiol Heart Circ Physiol 2023; 324:H762-H775. [PMID: 36930656 PMCID: PMC10151046 DOI: 10.1152/ajpheart.00638.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Plasma soluble prorenin receptor (sPRR) displays sexual dimorphism and is higher in women with type 2 diabetes mellitus (T2DM). However, the contribution of plasma sPRR to the development of vascular complications in T2DM remains unclear. We investigated if plasma sPRR contributes to sex differences in the activation of the systemic renin-angiotensin-aldosterone system (RAAS) and vascular damage in a model of high-fat diet (HFD)-induced T2DM. Male and female C57BL/6J mice were fed either a normal fat diet (NFD) or an HFD for 28 wk to assess changes in blood pressure, cardiometabolic phenotype, plasma prorenin/renin, sPRR, and ANG II. After completing dietary protocols, tissues were collected from males to assess vascular reactivity and aortic reactive oxygen species (ROS). A cohort of male mice was used to determine the direct contribution of increased systemic sPRR by infusion. To investigate the role of ovarian hormones, ovariectomy (OVX) was performed at 32 wk in females fed either an NFD or HFD. Significant sex differences were found after 28 wk of HFD, where only males developed T2DM and increased plasma prorenin/renin, sPRR, and ANG II. T2DM in males was accompanied by nondipping hypertension, carotid artery stiffening, and aortic ROS. sPRR infusion in males induced vascular thickening instead of material stiffening caused by HFD-induced T2DM. While intact females were less prone to T2DM, OVX increased plasma prorenin/renin, sPRR, and systolic blood pressure. These data suggest that sPRR is a novel indicator of systemic RAAS activation and reflects the onset of vascular complications during T2DM regulated by sex.NEW & NOTEWORTHY High-fat diet (HFD) for 28 wk leads to type 2 diabetes mellitus (T2DM) phenotype, concomitant with increased plasma soluble prorenin receptor (sPRR), nondipping blood pressure, and vascular stiffness in male mice. HFD-fed female mice exhibiting a preserved cardiometabolic phenotype until ovariectomy revealed increased plasma sPRR and blood pressure. Plasma sPRR may indicate the status of systemic renin-angiotensin-aldosterone system (RAAS) activation and the onset of vascular complications during T2DM in a sex-dependent manner.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
| | - Virginia Reverte
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Caleb M Abshire
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Carla B Rosales
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Michelle Galeas-Pena
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Nicholas R Harris
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Isabella Kilanowski-Doroh
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Margaret Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| |
Collapse
|
23
|
Longtine AG, Venkatasubramanian R, Zigler MC, Lindquist AJ, Mahoney SA, Greenberg NT, VanDongen NS, Ludwig KR, Moreau KL, Seals DR, Clayton ZS. Female C57BL/6N mice are a viable model of aortic aging in women. Am J Physiol Heart Circ Physiol 2023; 324:H893-H904. [PMID: 37115626 PMCID: PMC10202480 DOI: 10.1152/ajpheart.00120.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
The aorta stiffens with aging in both men and women, which predicts cardiovascular mortality. Aortic wall structural and extracellular matrix (ECM) remodeling, induced in part by chronic low-grade inflammation, contribute to aortic stiffening. Male mice are an established model of aortic aging. However, there is little information regarding whether female mice are an appropriate model of aortic aging in women, which we aimed to elucidate in the present study. We assessed two strains of mice and found that in C57BL/6N mice, in vivo aortic stiffness (pulse wave velocity, PWV) was higher with aging in both sexes, whereas in B6D2F1 mice, PWV was higher in old versus young male mice, but not in old versus young female mice. Because the age-related stiffening that occurs in men and women was reflected in male and female C57BL/6N mice, we examined the mechanisms of stiffening in this strain. In both sexes, aortic modulus of elasticity (pin myography) was lower in old mice, occurred in conjunction with and was related to higher plasma levels of the elastin-degrading enzyme matrix metalloproteinase-9 (MMP-9), and was accompanied by higher numbers of aortic elastin breaks and higher abundance of adventitial collagen-1. Plasma levels of the inflammatory cytokines interferon-γ, interleukin 6, and monocyte chemoattractant protein-1 were higher in both sexes of old mice. In conclusion, female C57BL/6N mice exhibit aortic stiffening, reduced modulus of elasticity and structural/ECM remodeling, and associated increases in MMP-9 and systemic inflammation with aging, and thus are an appropriate model of aortic aging in women.NEW & NOTEWORTHY Our study demonstrates that with aging, female C57BL/6N mice exhibit higher in vivo aortic stiffness, reduced modulus of elasticity, aortic wall structural and extracellular matrix remodeling, and elevations in systemic inflammation. These changes are largely reflective of those that occur with aging in women. Thus, female C57BL/6N mice are a viable model of human aortic aging and the utility of these animals should be considered in future biomedical investigations.
Collapse
Affiliation(s)
- Abigail G Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Melanie C Zigler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Alexandra J Lindquist
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nicholas S VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Katelyn R Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Kerrie L Moreau
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
24
|
Costa TJ, Barros PR, Duarte DA, Silva-Neto JA, Hott SC, Santos-Silva T, Costa-Neto CM, Gomes FV, Akamine EH, McCarthy CG, Jimenez-Altayó F, Dantas AP, Tostes RC. Carotid dysfunction in senescent female mice is mediated by increased α 1A-adrenoceptor activity and COX-derived vasoconstrictor prostanoids. Am J Physiol Heart Circ Physiol 2023; 324:H417-H429. [PMID: 36705993 PMCID: PMC11687965 DOI: 10.1152/ajpheart.00495.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/28/2023]
Abstract
α-Adrenergic receptors are crucial regulators of vascular hemodynamics and essential pharmacological targets for cardiovascular diseases. With aging, there is an increase in sympathetic activation, which could contribute to the progression of aging-associated cardiovascular dysfunction, including stroke. Nevertheless, there is little information directly associating adrenergic receptor dysfunction in the blood vessels of aged females. This study determined the role of a-adrenergic receptors in carotid dysfunction of senescent female mice (accelerated-senescence prone, SAMP8), compared with a nonsenescent (accelerated-senescence prone, SAMR1). Vasoconstriction to phenylephrine (Phe) was markedly increased in common carotid artery of SAMP8 [area under the curve (AUC), 527 ± 53] compared with SAMR1 (AUC, 334 ± 30, P = 0.006). There were no changes in vascular responses to the vasoconstrictor agent U46619 or the vasodilators acetylcholine (ACh) and sodium nitroprusside (NPS). Hyperactivity to Phe in female SAMP8 was reduced by cyclooxygenase-1 and cyclooxygenase-2 inhibition and associated with augmented ratio of TXA2/PGI2 release (SAMR1, 1.1 ± 0.1 vs. SAMP8, 2.1 ± 0.3, P = 0.007). However, no changes in cyclooxygenase expression were seen in SAMP8 carotids. Selective α1A-receptor antagonism markedly reduced maximal contraction, whereas α1D antagonism induced a minor shift in Phe contraction in SAMP8 carotids. Ligand binding analysis revealed a threefold increase of α-adrenergic receptor density in smooth muscle cells (VSMCs) of SAMP8 vs. SAMR1. Phe rapidly increased intracellular calcium (Cai2+) in VSMCs via the α1A-receptor, with a higher peak in VSMCs from SAMP8. In conclusion, senescence intensifies vasoconstriction mediated by α1A-adrenergic signaling in the carotid of female mice by mechanisms involving increased Cai2+ and release of cyclooxygenase-derived prostanoids.NEW & NOTEWORTHY The present study provides evidence that senescence induces hyperreactivity of α1-adrenoceptor-mediated contraction of the common carotid. Impairment of α1-adrenoceptor responses is linked to increased Ca2+ influx and release of COX-derived vasoconstrictor prostanoids, contributing to carotid dysfunction in the murine model of female senescence (SAMP8). Increased reactivity of the common carotid artery during senescence may lead to morphological and functional changes in arteries of the cerebral microcirculation and contribute to cognitive decline in females. Because the elderly population is growing, elucidating the mechanisms of aging- and sex-associated vascular dysfunction is critical to better direct pharmacological and lifestyle interventions to prevent cardiovascular risk in both sexes.
Collapse
Affiliation(s)
- Tiago J Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell Biology and Anatomy, Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, United States
| | - Paula R Barros
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Diego A Duarte
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Júlio A Silva-Neto
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sara Cristina Hott
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thamyris Santos-Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Claudio M Costa-Neto
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Eliana H Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Cameron G McCarthy
- Department of Cell Biology and Anatomy, Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, United States
| | - Francesc Jimenez-Altayó
- Department of Pharmacology, Therapeutic, and Toxicology, School of Medicine, Neuroscience Institute, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ana Paula Dantas
- Laboratory of Experimental Cardiology, Institut d'Investigacions Biomediques August Pi i Sunyer, Hospital Clinic Cardiovascular Institute, Barcelona, Spain
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
25
|
Sakamuri A, Ogola BO. The intersection between senescence-mediated vascular dysfunction and cognitive impairment in female mice. Am J Physiol Heart Circ Physiol 2023; 324:H411-H413. [PMID: 36800510 PMCID: PMC9988518 DOI: 10.1152/ajpheart.00076.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Affiliation(s)
- Anil Sakamuri
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Bernard Ojwang Ogola
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
26
|
Wnuk A, Przepiórska K, Pietrzak BA, Kajta M. Emerging Evidence on Membrane Estrogen Receptors as Novel Therapeutic Targets for Central Nervous System Pathologies. Int J Mol Sci 2023; 24:ijms24044043. [PMID: 36835454 PMCID: PMC9968034 DOI: 10.3390/ijms24044043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Nuclear- and membrane-initiated estrogen signaling cooperate to orchestrate the pleiotropic effects of estrogens. Classical estrogen receptors (ERs) act transcriptionally and govern the vast majority of hormonal effects, whereas membrane ERs (mERs) enable acute modulation of estrogenic signaling and have recently been shown to exert strong neuroprotective capacity without the negative side effects associated with nuclear ER activity. In recent years, GPER1 was the most extensively characterized mER. Despite triggering neuroprotective effects, cognitive improvements, and vascular protective effects and maintaining metabolic homeostasis, GPER1 has become the subject of controversy, particularly due to its participation in tumorigenesis. This is why interest has recently turned toward non-GPER-dependent mERs, namely, mERα and mERβ. According to available data, non-GPER-dependent mERs elicit protective effects against brain damage, synaptic plasticity impairment, memory and cognitive dysfunctions, metabolic imbalance, and vascular insufficiency. We postulate that these properties are emerging platforms for designing new therapeutics that may be used in the treatment of stroke and neurodegenerative diseases. Since mERs have the ability to interfere with noncoding RNAs and to regulate the translational status of brain tissue by affecting histones, non-GPER-dependent mERs appear to be attractive targets for modern pharmacotherapy for nervous system diseases.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Correspondence: (A.W.); (M.K.); Tel.: +48-12-662-3339 (A.W.); +48-12-662-3235 (M.K.); Fax: +48-12-637-4500 (A.W. & M.K.)
| | | | | | - Małgorzata Kajta
- Correspondence: (A.W.); (M.K.); Tel.: +48-12-662-3339 (A.W.); +48-12-662-3235 (M.K.); Fax: +48-12-637-4500 (A.W. & M.K.)
| |
Collapse
|
27
|
Horton AC, Wilkinson MM, Kilanowski-Doroh I, Ogola BO, Lindsey SH. Dihydrotestosterone Induces Arterial Stiffening in Female Mice. RESEARCH SQUARE 2023:rs.3.rs-2522089. [PMID: 36798163 PMCID: PMC9934771 DOI: 10.21203/rs.3.rs-2522089/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Background Testosterone is the predominant sex hormone in men and is increased in women with polycystic ovarian syndrome. These patients also experience an increased risk for cardiovascular diseases including hypertension and arterial stiffness. Since our previous work shows an important role for the G protein-coupled estrogen receptor (GPER) in arterial stiffness, we hypothesized that other hormones including androgens may impact arterial stiffness in female mice via regulation of GPER. Methods The impact of the non-aromatizable androgen dihydrotestosterone (DHT), the glucocorticoid dexamethasone, and the progestin medroxyprogesterone acetate (all 100 nM for 24 h) on GPER and ERα expression was assessed in cultured vascular smooth muscle cells using droplet digital PCR (ddPCR). To assess the in vivo impact of the DHT-induced downregulation of GPER, female ovary-intact C57Bl/6 mice were treated with silastic capsules containing DHT for 4 weeks, one with a dosage expected to mimic human male DHT levels and another to double the expected human concentration (n=8-9/group). Results GPER mRNA was only decreased by DHT (P=0.001), while ERα expression was significantly suppressed by all hormones (P<0.0001). While blood pressure was not different between groups (P= 0.59), there was a dose-dependent increase in body weight (control 22±2 g, single dose 24±2 g, double dose 26±2 g; P=0.0002). Intracarotid stiffness measured via pulse wave velocity showed a more than two-fold increase in both DHT-treated groups (control 1.9±0.3 m/s, single dose 4.3±0.8 m/s, double dose 4.8±1.0 m/s). Histological analysis of aortic sections using Masson's trichrome showed a significant decrease in collagen between the control group (24 ± 5%) and the double dose group (17 ± 3%, P=0.007), despite no changes in aortic wall thickness or smooth muscle content. Lastly, ddPCR showed that in vivo DHT treatment decreased aortic expression of both GPER (control 20±5, single dose 10.5 ± 5.6, double dose 10±4 copies/ng; P=0.001) and ERα (control 54±2, single dose 24±13, and double dose 23 ± 12 copies/ng; P=0.003). Conclusions These findings indicate that testosterone promotes arterial stiffening and cardiovascular damage in female mice and is associated with decreased estrogen receptor expression. These data are important not only for polycystic ovarian syndrome patients but also women using testosterone for fitness, gender transitioning, or reduced libido.
Collapse
Affiliation(s)
- Alec C Horton
- Tulane University Department of Pharmacology and the Tulane Brain Institute, New Orleans, LA
| | | | | | - Benard O Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA
| | - Sarah H Lindsey
- Tulane University Department of Pharmacology, the Tulane Brain Institute, and the Tulane Center of Excellence in Sex-Based Biology and Medicine, New Orleans, LA
| |
Collapse
|
28
|
Kehmeier MN, Bedell BR, Cullen AE, Khurana A, D'Amico HJ, Henson GD, Walker AE. In vivo arterial stiffness, but not isolated artery endothelial function, varies with the mouse estrous cycle. Am J Physiol Heart Circ Physiol 2022; 323:H1057-H1067. [PMID: 36240435 PMCID: PMC9678414 DOI: 10.1152/ajpheart.00369.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022]
Abstract
With the increasing appreciation for sex as a biological variable and the inclusion of female mice in research, it is important to understand the influence of the estrous cycle on physiological function. Sex hormones are known to modulate vascular function, but the effects of the mouse estrous cycle phase on arterial stiffness, endothelial function, and arterial estrogen receptor expression remain unknown. In 23 female C57BL/6 mice (6 mo of age), we determined the estrous cycle stage via vaginal cytology and plasma hormone concentrations. Aortic stiffness, assessed by pulse wave velocity, was lower during the estrus phase compared with diestrus. In ex vivo assessment of isolated pressurized mesenteric and posterior cerebral arteries, the responses to acetylcholine, insulin, and sodium nitroprusside, as well as nitric oxide-mediated dilation, were not different between estrous cycle phases. In the aorta, expression of phosphorylated estrogen receptor-α was higher for mice in estrus compared with mice in proestrus. In the cerebral arteries, gene expression for estrogen receptor-β (Esr2) was lowest for mice in estrus compared with diestrus and proestrus. These results demonstrate that the estrus phase is associated with lower in vivo large artery stiffness in mice. In contrast, ex vivo resistance artery endothelial function is not different between estrous cycle phases. Estrogen receptor expression is modulated by the estrus cycle in an artery-dependent manner. These results suggest that the estrous cycle phase should be considered when measuring in vivo arterial stiffness in young female mice.NEW & NOTEWORTHY To design rigorous vascular research studies using young female rodents, the influence of the estrous cycle on vascular function must be known. We found that in vivo aortic stiffness was lower during estrus compared with the diestrus phase in female mice. In contrast, ex vivo mesenteric and cerebral artery endothelial function did not differ between estrous cycle stages. These results suggest that the estrous cycle stage should be accounted for when measuring in vivo arterial stiffness.
Collapse
Affiliation(s)
| | - Bradley R Bedell
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Abigail E Cullen
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Aleena Khurana
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Holly J D'Amico
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Grant D Henson
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|