1
|
Aloisi AM, Casini I. Fibromyalgia: Chronic Pain Due to a Blood Dysfunction? Int J Mol Sci 2025; 26:4153. [PMID: 40362392 PMCID: PMC12071621 DOI: 10.3390/ijms26094153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Fibromyalgia (FM) is a common chronic disorder with chronic pain. FM generally affects all ages and occurs more commonly in women. The cause of FM remains undefined, but a number of factors suggest the cardiovascular system and the blood in particular as contributors to its occurrence and maintenance. Hemograms and other blood indexes often show high percentages of values at the 'normal', low, or high limits and several values outside of the 'normal' ranges. On the other hand, vessels regulate blood arrival to tissues depending on many internal and external factors. Both aspects can interfere with tissue oxygenation and then with the numerous consequences induced by hypoxia. In this narrative review, efforts were made to highlight factors that are potentially able to affect oxygen arrival in cells, as well as other factors related to blood elements that can play a role in the chronic pain experienced by FM patients. Data strongly indicate that most of the symptoms commonly present in FM patients can find their physio-pathological basis in the blood, suggesting blood-related interventions in these patients.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- Stress and Pain Neurophysiology Laboratory, Department of Medicine, Surgery and Neuroscience University of Siena, 53100 Siena, Italy;
| | | |
Collapse
|
2
|
Ramsay Z, Sharma D, Wisdom-Phipps M, Chin N, Campbell L, Knight-Madden J, Asnani M. Estradiol is Pro-Nociceptive and Associated with a Small-Fiber Neuropathy Among Premenopausal Women with Sickle Cell Disease. Hemoglobin 2025:1-11. [PMID: 40230074 DOI: 10.1080/03630269.2025.2489634] [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: 02/18/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Vaso-occlusive crisis (VOC) pain episodes often occur with menses and concurrent hormonal contraceptive use may reduce their frequency. This study tested the hypothesis that among women with sickle cell disease (SCD), sex hormones are associated with pain detection thresholds. Women with SCD aged minimum 18 years with regular menses, and without acute illnesses, pregnancy, oophorectomy, or hormonal contraceptive use within three months prior were included. Pain detection thresholds for heat (HPT) and pressure (PPT), and serum estradiol, progesterone and testosterone were measured at follicular and luteal phases. The Adult Sickle Cell Quality-of-Life Measurement Information System assessed quality-of-life and VOC frequency and severity scores. Generalized linear mixed models were performed, including the day of the cycle standardized by cycle length. Among 125 participants, neither the day nor phase of the menstrual cycle was associated with PPT or HPT. In multivariate analyses, worse VOC scores (β = 1.7) and severe genotype (β = -46.0) were associated with higher and lower trapezius PPT, respectively. Older age was associated with lower forearm HPT (β = -0.1). Among leg measurements, ovulatory cycles (β = -1.1) and hydroxyurea use (β = -1.2) were associated with lower HPT, while worse VOC scores (β = 0.1) were associated with higher HPT. Higher estradiol was associated with lower HPT at the leg (β = -0.02), with an interaction with the cycle day (β = 0.001) predicting lower thresholds earlier in the cycle for the same estradiol level. Estradiol is associated with a time-varying, length-dependent small-fiber neuropathy among SCD women; and may be a potential therapeutic target and biomarker for SCD pain.
Collapse
Affiliation(s)
- Zachary Ramsay
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| | - Deva Sharma
- Division of Transfusion Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret Wisdom-Phipps
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| | - Nicki Chin
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| | - Leroy Campbell
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| | - Jennifer Knight-Madden
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| | - Monika Asnani
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies Mona Campus, Kingston 7, Jamaica
| |
Collapse
|
3
|
Valenzuela-Fuenzalida JJ, Núñez-Castro CI, Morán-Durán VB, Nova-Baeza P, Orellana-Donoso M, Suazo-Santibáñez A, Becerra-Farfan A, Oyanedel-Amaro G, Bruna-Mejias A, Granite G, Casanova-Martinez D, Sanchis-Gimeno J. Anatomical Variants in Pancreatic Irrigation and Their Clinical Considerations for the Pancreatic Approach and Surrounding Structures: A Systematic Review with Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:666. [PMID: 40282957 PMCID: PMC12028877 DOI: 10.3390/medicina61040666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: The pancreas receives blood through a complex network of multiple branches, primarily originating from the celiac trunk (CeT) and the superior mesenteric artery (SMA). This blood supply is structured into three main arterial groups, each serving different regions of the pancreas to effectively support its endocrine and exocrine functions. Materials and Methods: The databases Medline, Scopus, Web of Science, Google Scholar, Cumulative Index to Nursing and Allied Health Literature (CINAHL) and Latin American and the Caribbean Literature in Health Sciences (LILACS) were searched until January 2025. Methodological quality was evaluated using an assurance tool for anatomical studies (AQUA). Pooled prevalence was estimated using a random effects model. Results: A total of sixteen studies met the established selection criteria in this study for meta-analysis. Pancreatic irrigation variants presented a prevalence of 11.2% (CI: 7-14%) and a heterogeneity of 88.2%. The other studies were analyzed by subgroups, showing statistically significant differences in the following subgroups: (1) sample type-a larger sample of images analyzed in the included studies (p = 0.312), which did not show statistically significant differences; (2) geographical region (p = 0.041), which showed a greater presence in the Asian population studied, and this was statistically significant; and (3) sex (male or female) (p = 0.12), where there were no statistically significant differences. Conclusions: The discovery of variations in pancreatic irrigation is common due to the numerous blood vessels involved in supplying this vital organ. Understanding different vascular patterns (such as those from the splenic and mesenteric arteries) is crucial for surgical interventions on the pancreas. For transplant patients, a thorough vascular analysis of both the donor and recipient is essential. Variations can impact blood flow and compatibility, potentially leading to transplant rejection if not addressed. To enhance outcomes, it is recommended to develop more accurate imaging tools for pre-surgical analysis, necessitating ongoing research in this area.
Collapse
Affiliation(s)
- Juan José Valenzuela-Fuenzalida
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Camila Ignacia Núñez-Castro
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Valeria Belén Morán-Durán
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Pablo Nova-Baeza
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Mathias Orellana-Donoso
- Escuela de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile;
- Department of Morphological Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510157, Chile
| | | | - Alvaro Becerra-Farfan
- Departamento de Ciencias Química y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370993, Chile;
| | - Gustavo Oyanedel-Amaro
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| | - Alejandro Bruna-Mejias
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
- Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso 2360072, Chile
| | - Guinevere Granite
- Department of Surgery, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, ML 20814, USA;
| | - Daniel Casanova-Martinez
- Facultad de Medicina, Universidad de Valparaíso, Campus San Felipe, Valparaíso 2170000, Chile;
- Laboratorio de Neuroanatomía Microquirúrgica (LaNeMic), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1053, Argentina
| | - Juan Sanchis-Gimeno
- GIAVAL Research Group, Department of Anatomy and Human Embryology, Faculty of Medicine, University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
4
|
Addisu D, Mitiku Y, Yazie Ferede W, Mekuriaw BY, Erega BB, Bazezew LY, Belachew TW, Tadesse SG, Goshu YA, Mengistie BA, Mekie M, Misker AD, Mihretie GN. Determinants of new-onset postpartum preeclampsia among mothers who delivered in hospitals in the South Gondar Zone, Northwest Ethiopia: a multicenter case-control study. BMC Pregnancy Childbirth 2025; 25:161. [PMID: 39953424 PMCID: PMC11829449 DOI: 10.1186/s12884-025-07274-z] [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: 12/25/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND New-onset postpartum preeclampsia has emerging as a significant public health concern in Ethiopia, with a notable increase in incidence in the study area. While substantial research exists on antepartum preeclampsia, data on the determinants of new-onset postpartum preeclampsia are limited. Therefore, this study aimed to investigate determinants of new-onset postpartum preeclampsia among mothers who gave birth at hospitals in the South Gondar Zone, Northwest Ethiopia. METHODS An unmatched case-control study was conducted with 355 postpartum mothers (89 cases and 266 controls). A multistage sampling method was used to select the study participants. Data were collected using structured questionnaires and medical chart reviews, and analyzed using SPSS Version 27. Binary logistic regression (bivariable and multivariable analysis) was used to identify factors associated with new-onset postpartum preeclampsia. Statistical significance was set at p < 0.05, and strength of associations were measured using adjusted odds ratios (AOR) with 95% confidence intervals (CIs). RESULT New-onset postpartum preeclampsia was significantly associated with several factors, including advanced maternal age, fewer antenatal care (ANC) visits, contraceptive use, physical inactivity, a history of multiple gestations, and gestational diabetes mellitus. Specifically, maternal age ≥ 40 years (AOR = 11.63, 95% CI: 4.24-31.86), fewer than four ANC visits (AOR = 8.45, 95% CI: 3.96-18.05), contraceptive use (AOR = 4.04, 95% CI: 1.26-13.37), irregular physical activity (AOR = 4.05, 95% CI: 1.32-12.44), physical inactivity (AOR = 8.25, 95% CI: 4.62-19.29), a history of multiple gestations (AOR = 2.66, 95% CI: 1.26-5.60), and gestational diabetes mellitus (AOR = 17.79, 95% CI: 7.72-40.95) were identified as key determinants. CONCLUSIONS Advanced maternal age, fewer ANC visits, contraceptive use, physical inactivity, multiple gestations, and gestational diabetes were strongly associated with new-onset postpartum preeclampsia. Increased ANC visits and postpartum monitoring are essential for early detection and management of postpartum preeclampsia. Additionally, promoting physical activity should be incorporated into maternal health strategies to reduce the incidence of new onset postpartum preeclampsia.
Collapse
Affiliation(s)
- Dagne Addisu
- Department of Midwifery, Debre Tabor University, Debre Tabor, Ethiopia.
| | - Yekaba Mitiku
- Department of Midwifery, Debre Tabor Comprehensive Specialized Hospital, Debre Tabor, Ethiopia
| | | | | | | | - Lakachew Yismaw Bazezew
- Department of Pediatrics and Neonatal Nursing, Debre Tabor University, Debre Tabor, Ethiopia
| | | | | | | | | | - Maru Mekie
- Department of Midwifery, Debre Tabor University, Debre Tabor, Ethiopia
| | | | | |
Collapse
|
5
|
Ribeiro Reis AP, Ioannidou E, Stuart KV, Wagner SK, Foster PJ, Khawaja AP, Petzold A, Sivaprasad S, Pontikos N, Keane PA, Balaskas K, Patel PJ. Macular, choroidal and disc associations across women's reproductive life stages: a scoping review from menarche to post-menopause. Eye (Lond) 2025; 39:402-411. [PMID: 39814872 PMCID: PMC11794579 DOI: 10.1038/s41433-025-03592-w] [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: 04/22/2024] [Revised: 11/17/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
Oestrogen and progesterone fluctuate cyclically in women throughout their adult lives. Although these hormones cross the blood-retinal barrier and bind to intraocular receptors, their effects remain unclear. We present the first review to date on associations between posterior pole structures-specifically the macula, choroid, and optic disc-and both the menstrual cycle and post-menopausal period, utilising multimodal imaging techniques in healthy adult non-pregnant women. We excluded studies on contraception and hormonal replacement therapy, focusing solely on physiological associations. Despite the comprehensive scope of our review, limited data and inconsistent reporting among studies prevented the establishment of meaningful trends. Across menstrual cycle phases, choroidal thickness (CHT) was the most consistently reported parameter, with thinning during the luteal phase compared to the follicular phase. Conversely, no significant differences were observed in macular or disc morphology across the cycle, likely reflecting a preserved structure despite potential fluctuations in blood flow and perfusion. Studies comparing pre- and post-menopausal associations, after adjusting for age or body mass index (BMI), failed to reveal meaningful trends, highlighting the difficulty in separating the effect of age from hormonal declines in older women. Understanding how hormonal cycles impact the posterior pole in women is crucial for addressing sex differences in various ocular pathologies. Research on female-specific factors is still sparse, and interestingly, the majority of affiliations in the reviewed articles did not originate from regions with the highest biomedical research funding and publication rates. We encourage further studies focusing on female-specific variables and provide recommendations for future designs.
Collapse
Affiliation(s)
- Ana Paula Ribeiro Reis
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.
| | - Estelle Ioannidou
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Kelsey V Stuart
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Siegfried K Wagner
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Paul J Foster
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Anthony P Khawaja
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Axel Petzold
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Sobha Sivaprasad
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Nikolas Pontikos
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Pearse A Keane
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Konstantinos Balaskas
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Praveen J Patel
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
6
|
Azzam AY, Morsy MM, Ellabban MH, Morsy AM, Zahran AA, Nassar M, Elsayed OS, Elswedy A, Elamin O, Al Zomia AS, Abukhadijah HJ, Alotaibi HA, Atallah O, Azab MA, Essibayi MA, Dmytriw AA, Morsy MD, Altschul DJ. The Impact of Idiopathic Intracranial Hypertension on Cardiovascular Disease Risk Among UK Women: An Obesity-Adjusted Analysis. ASIDE INTERNAL MEDICINE 2025; 1:1-11. [PMID: 39830613 PMCID: PMC11739732 DOI: 10.71079/h1fr8h68] [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: 01/22/2025]
Abstract
Introduction Idiopathic intracranial hypertension (IIH) is known to elevate cardiovascular disease (CVD) risk, but the extent to which obesity and IIH-specific factors contribute to this risk is not well understood. WE aim to separate the effects of obesity from IIH-specific factors on the risk of stroke and CVD, building on previous findings that indicate a two-fold increase in cardiovascular events in women with IIH compared to BMI-matched controls. Methods An obesity-adjusted risk analysis was conducted using Indirect Standardization based on data from a cohort study by Adderley et al., which included 2,760 women with IIH and 27,125 matched healthy controls from The Health Improvement Network (THIN). Advanced statistical models were employed to adjust for confounding effects of obesity and determine the risk contributions of IIH to ischemic stroke and CVD, independent of obesity. Four distinct models explored the interactions between IIH, obesity, and CVD risk. Results The analysis showed that IIH independently contributes to increased cardiovascular risk beyond obesity alone. Risk ratios for cardiovascular outcomes were significantly higher in IIH patients compared to controls within similar obesity categories. Notably, a synergistic effect was observed in obese IIH patients, with a composite CVD risk ratio of 6.19 (95% CI: 4.58-8.36, p<0.001) compared to non-obese controls. Conclusions This study underscores a significant, independent cardiovascular risk from IIH beyond obesity. The findings advocate for a shift in managing IIH to include comprehensive cardiovascular risk assessment and mitigation. Further research is required to understand the mechanisms and develop specific interventions for this group.
Collapse
Affiliation(s)
- Ahmed Y. Azzam
- Faculty of Medicine, October 6 University, 6
of October City, Giza, Egypt
- Montefiore-Einstein Cerebrovascular Research Lab, Albert
Einstein College of Medicine, Bronx, NY, USA
- Director of Clinical Research and Clinical Artificial
Intelligence, American Society for Inclusion, Diversity, and Health Equity (ASIDE),
Delaware, USA
| | - Mahmoud M. Morsy
- Faculty of Medicine, October 6 University, 6
of October City, Giza, Egypt
- Clinical Research Fellow, American Society for Inclusion,
Diversity, and Health Equity (ASIDE), Delaware, USA
| | | | - Ahmed M. Morsy
- Kasr Alainy Faculty of Medicine, Cairo University
Hospitals, Cairo University, Cairo, Egypt
| | - Adham Adel Zahran
- Kasr Alainy Faculty of Medicine, Cairo University
Hospitals, Cairo University, Cairo, Egypt
| | - Mahmoud Nassar
- Department of Medicine, Division of Endocrinology,
Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences,
University at Buffalo, New York, USA
- Founder, American Society for Inclusion, Diversity, and
Health Equity (ASIDE), Delaware, USA
| | - Omar S. Elsayed
- Faculty of Medicine, October 6 University, 6
of October City, Giza, Egypt
| | - Adam Elswedy
- Faculty of Medicine, October 6 University, 6
of October City, Giza, Egypt
| | - Osman Elamin
- Department of Neurosurgery, Jordan Hospital, Amman,
Jordan
| | | | | | - Hammam A. Alotaibi
- Ophthalmology Department, Prince Sultan Military Medical
City, Riyadh, Saudi Arabia
| | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School,
Hannover, Germany
| | - Mohammed A. Azab
- Department of Neurosurgery, Cleveland Clinic Foundation,
Cleveland, OH, USA
| | - Muhammed Amir Essibayi
- Montefiore-Einstein Cerebrovascular Research Lab, Albert
Einstein College of Medicine, Bronx, NY, USA
- Department of Neurological Surgery, Montefiore Medical
Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adam A. Dmytriw
- Neuroendovascular Program, Massachusetts General
Hospital & Brigham and Women’s Hospital, Harvard University, Boston, MA,
USA
- Neurovascular Centre, Divisions of Therapeutic
Neuroradiology & Neurosurgery, St. Michael’s Hospital, University of
Toronto, Toronto, ON, Canada
| | | | - David J. Altschul
- Montefiore-Einstein Cerebrovascular Research Lab, Albert
Einstein College of Medicine, Bronx, NY, USA
- Department of Neurological Surgery, Montefiore Medical
Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
7
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
8
|
Meegaswatte H, Speer K, McKune AJ, Naumovski N. Functional Foods and Nutraceuticals for the Management of Cardiovascular Disease Risk in Postmenopausal Women. Rev Cardiovasc Med 2024; 25:460. [PMID: 39742223 PMCID: PMC11683719 DOI: 10.31083/j.rcm2512460] [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: 05/16/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 01/03/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death in women and risk of development is greatly increased following menopause. Menopause occurs over several years and is associated with hormonal changes, including a reduction in estradiol and an increase in follicle-stimulating hormone. This hormonal shift may result in an increased risk of developing abdominal adiposity, insulin resistance, dyslipidemia, vascular dysfunction, hypertension, type 2 diabetes mellitus (T2DM), metabolic dysfunction-associated fatty liver disease (MAFLD), and metabolic syndrome (MetS). Furthermore, with the onset of menopause, there is an increase in oxidative stress that is associated with impaired vascular function, inflammation, and thrombosis, further increasing the risk of CVD development. Despite the harmful consequences of the menopause transition being well known, women in premenopausal, perimenopausal, and postmenopausal stages are unlikely to be enrolled in research studies. Therefore, investigations on the prevention and treatment of cardiovascular and metabolic disease in middle-aged women are still relatively limited. Whilst lifestyle interventions are associated with reduced CVD risk in this population sample, the evidence still remains inconclusive. Therefore, it is important to explore the effectiveness of early intervention and potential therapeutic approaches to maintain cellular redox balance, preserve endothelium, and reduce inflammation. Glycine, N-acetylcysteine, and L-theanine are amino acids with potential antioxidant and anti-inflammatory activity and are identified as therapeutic interventions in the management of age-related and metabolic diseases. The benefits of the intake of these amino acids for improving factors associated with cardiovascular health are discussed in this review. Future studies using these amino acids are warranted to investigate their effect on maintaining the vascular health and cardiovascular outcomes of postmenopausal women.
Collapse
Affiliation(s)
- Harshini Meegaswatte
- Faculty of Health, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- University of Canberra Research Institute for Sport and Exercise (UCRISE), University of Canberra, 2617 Bruce, Canberra, ACT, Australia
| | - Kathryn Speer
- Faculty of Health, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- University of Canberra Research Institute for Sport and Exercise (UCRISE), University of Canberra, 2617 Bruce, Canberra, ACT, Australia
| | - Andrew J. McKune
- Faculty of Health, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- University of Canberra Research Institute for Sport and Exercise (UCRISE), University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Discipline of Biokinetics, Exercise and Leisure Sciences, School of Health Science, University of KwaZulu-Natal, 4041 Durban, Republic of South Africa
| | - Nenad Naumovski
- Faculty of Health, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- University of Canberra Research Institute for Sport and Exercise (UCRISE), University of Canberra, 2617 Bruce, Canberra, ACT, Australia
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece
| |
Collapse
|
9
|
Ribeiro Reis AP, Ioannidou E, Wagner SK, Struyven R, Sun Z, Foster P, Khawaja AP, Petzold A, Sivaprasad S, Pontikos N, Keane PA, Balaskas K, Greco E, Iliodromiti S, Patel PJ. Retinal morphology across the menstrual cycle: insights from the UK Biobank. NPJ WOMEN'S HEALTH 2024; 2:38. [PMID: 39654609 PMCID: PMC11627222 DOI: 10.1038/s44294-024-00042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024]
Abstract
Oestradiol and progesterone levels are higher in menstruating women than men of the same age, and their receptors are present in their neurosensory retina and retinal pigment epithelium. However, the impact of this hormonal environment on retinal physiology in women remains unclear. Using self-reported menstrual cycle phases as a surrogate for fluctuating hormonal levels, we investigated associations with retinovascular indices on colour fundus photograph and retinal thickness in optical coherence tomography across regularly menstruating women in the UK Biobank. We found no differences in retinal thickness across the cycle; however, vessel density, arteriolar and venular, and fractal dimension were higher in the luteal phase than follicular. The calibre of the central retinal vessels did not differ. This study suggests that the menstrual cycle phase might be associated with retinal microvasculature density in non-invasive imaging. It raises awareness for this understudied area, providing insights into neuroscience fields and epidemiological studies.
Collapse
Affiliation(s)
- Ana Paula Ribeiro Reis
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Estelle Ioannidou
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Siegfried Karl Wagner
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Robbert Struyven
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
- Centre for Medical Image Computing, University College London, London, UK
| | - Zihan Sun
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Paul Foster
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Anthony P. Khawaja
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Axel Petzold
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Sobha Sivaprasad
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Nikolas Pontikos
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Pearse A. Keane
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Konstantinos Balaskas
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Elena Greco
- The Royal London Hospital, Barts Health NHS Trust, London, UK
| | | | - Praveen J. Patel
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
10
|
Carlström M, Weitzberg E, Lundberg JO. Nitric Oxide Signaling and Regulation in the Cardiovascular System: Recent Advances. Pharmacol Rev 2024; 76:1038-1062. [PMID: 38866562 DOI: 10.1124/pharmrev.124.001060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Nitric oxide (NO) from endothelial NO synthase importantly contributes to vascular homeostasis. Reduced NO production or increased scavenging during disease conditions with oxidative stress contribute to endothelial dysfunction and NO deficiency. In addition to the classical enzymatic NO synthases (NOS) system, NO can also be generated via the nitrate-nitrite-NO pathway. Dietary and pharmacological approaches aimed at increasing NO bioactivity, especially in the cardiovascular system, have been the focus of much research since the discovery of this small gaseous signaling molecule. Despite wide appreciation of the biological role of NOS/NO signaling, questions still remain about the chemical nature of NOS-derived bioactivity. Recent studies show that NO-like bioactivity can be efficiently transduced by mobile NO-ferroheme species, which can transfer between proteins, partition into a hydrophobic phase, and directly activate the soluble guanylyl cyclase-cGMP-protein kinase G pathway without intermediacy of free NO. Moreover, interaction between red blood cells and the endothelium in the regulation of vascular NO homeostasis have gained much attention, especially in conditions with cardiometabolic disease. In this review we discuss both classical and nonclassical pathways for NO generation in the cardiovascular system and how these can be modulated for therapeutic purposes. SIGNIFICANCE STATEMENT: After four decades of intensive research, questions persist about the transduction and control of nitric oxide (NO) synthase bioactivity. Here we discuss NO signaling in cardiovascular health and disease, highlighting new findings, such as the important role of red blood cells in cardiovascular NO homeostasis. Nonclassical signaling modes, like the nitrate-nitrite-NO pathway, and therapeutic opportunities related to the NO system are discussed. Existing and potential pharmacological treatments/strategies, as well as dietary components influencing NO generation and signaling are covered.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| |
Collapse
|
11
|
Xega V, Liu JL. Beyond reproduction: unraveling the impact of sex hormones on cardiometabolic health. MEDICAL REVIEW (2021) 2024; 4:284-300. [PMID: 39135604 PMCID: PMC11317208 DOI: 10.1515/mr-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/07/2024] [Indexed: 08/15/2024]
Abstract
This review thoroughly explores the multifaceted roles of sexual hormones, emphasizing their impact beyond reproductive functions and underscoring their significant influence on cardiometabolic regulation. It analyzes the broader physiological implications of estrogen, testosterone, and progesterone, highlighting their effects on metabolic syndrome, lipid metabolism, glucose homeostasis, and cardiovascular health. Drawing from diverse molecular, clinical, and therapeutic studies, the paper delves into the intricate interplay between these hormones and cardiometabolic processes. By presenting a comprehensive analysis that goes beyond traditional perspectives, and recognizing sexual hormones as more than reproductive agents, the review sheds light on their broader significance in health and disease management, advocating for holistic and personalized medical approaches.
Collapse
Affiliation(s)
- Viktoria Xega
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Jun-Li Liu
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
12
|
Corona G, Rastrelli G, Bianchi N, Sparano C, Sforza A, Vignozzi L, Maggi M. Hyperprolactinemia and male sexual function: focus on erectile dysfunction and sexual desire. Int J Impot Res 2024; 36:324-332. [PMID: 37340146 DOI: 10.1038/s41443-023-00717-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 06/22/2023]
Abstract
The present paper aims to analyze and discuss the available evidence supporting the relationship between male sexual function and elevated prolactin (PRL) levels (HPRL). Two different sources of data were analyzed. Clinical data were derived from a series of patients seeking medical care for sexual dysfunction at our Unit. Out of 418 studies, 25 papers were used with a meta-analytic approach to evaluate the overall prevalence of HPRL in patients with erectile dysfunction (ED) and to study the influence of HPRL and its treatment on male sexual function. Among 4215 patients (mean age 51.6 ± 13.1 years) consulting for sexual dysfunction at our Unit, 176 (4.2%) showed PRL levels above the normal range. Meta-analytic data showed that HPRL is a rare condition among patients with ED (2 [1;3]%). Either clinical and meta-analytic data confirm a stepwise negative influence of PRL on male sexual desire (S = 0.00004 [0.00003; 0.00006]; I = -0.58915 [-0.78438; -0.39392]; both p < 0.0001 from meta-regression analysis). Normalization of PRL levels is able to improve libido. The role of HPRL in ED remains inconclusive. Data from a meta-analytic approach showed that either HPRL or reduced T levels were independently associated with ED rates. The normalization of PRL levels only partially restored ED. HPRL did not significantly contribute to ED severity, in our clinical setting. In conclusion, treating HPRL can restore normal sexual desire, whereas its effect on erection is limited.
Collapse
Affiliation(s)
- G Corona
- Endocrinology Unit, Maggiore-Bellaria Hospital, Medical Department, Azienda-Usl Bologna, Bologna, Italy.
| | - G Rastrelli
- Andrology, Female Endocrinology and Gender Incongruence Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - N Bianchi
- Endocrinology Unit, Maggiore-Bellaria Hospital, Medical Department, Azienda-Usl Bologna, Bologna, Italy
| | - C Sparano
- Endocrinology Unit Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - A Sforza
- Endocrinology Unit, Maggiore-Bellaria Hospital, Medical Department, Azienda-Usl Bologna, Bologna, Italy
| | - L Vignozzi
- Andrology, Female Endocrinology and Gender Incongruence Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - M Maggi
- Endocrinology Unit Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Li C, Ajmal E, Alok K, Powell K, Wadolowski S, Tambo W, Turpin J, Barthélemy E, Al-Abed Y, LeDoux D. CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury. Biol Sex Differ 2024; 15:44. [PMID: 38816868 PMCID: PMC11138127 DOI: 10.1186/s13293-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression. METHODS Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes. RESULTS Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males. CONCLUSIONS This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.
Collapse
Affiliation(s)
- Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA.
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Khaled Alok
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Steven Wadolowski
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Justin Turpin
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Ernest Barthélemy
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - David LeDoux
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| |
Collapse
|
14
|
Nasr MK, Schurmann C, Böttinger EP, Teumer A. Mendelian randomization indicates causal effects of estradiol levels on kidney function in males. Front Endocrinol (Lausanne) 2023; 14:1232266. [PMID: 38169598 PMCID: PMC10758447 DOI: 10.3389/fendo.2023.1232266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Context Chronic kidney disease (CKD) is a public health burden worldwide. Epidemiological studies observed an association between sex hormones, including estradiol, and kidney function. Objective We conducted a Mendelian randomization (MR) study to assess a possible causal effect of estradiol levels on kidney function in males and females. Design We performed a bidirectional two-sample MR using published genetic associations of serum levels of estradiol in men (n = 206,927) and women (n = 229,966), and of kidney traits represented by estimated glomerular filtration rate (eGFR, n = 567,460), urine albumin-to-creatinine ratio (UACR, n = 547,361), and CKD (n = 41,395 cases and n = 439,303 controls) using data obtained from the CKDGen Consortium. Additionally, we conducted a genome-wide association study using UK Biobank cohort study data (n = 11,798 men and n = 6,835 women) to identify novel genetic associations with levels of estradiol, and then used these variants as instruments in a one-sample MR. Results The two-sample MR indicated that genetically predicted estradiol levels are significantly associated with eGFR in men (beta = 0.077; p = 5.2E-05). We identified a single locus at chromosome 14 associated with estradiol levels in men being significant in the one-sample MR on eGFR (beta = 0.199; p = 0.017). We revealed significant results with eGFR in postmenopausal women and with UACR in premenopausal women, which did not reach statistical significance in the sensitivity MR analyses. No causal effect of eGFR or UACR on estradiol levels was found. Conclusions We conclude that serum estradiol levels may have a causal effect on kidney function. Our MR results provide starting points for studies to develop therapeutic strategies to reduce kidney disease.
Collapse
Affiliation(s)
- M. Kamal Nasr
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Claudia Schurmann
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
| | - Erwin P. Böttinger
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
15
|
Al-Hassany L, Linstra KM, Meun C, van den Berg J, Boersma E, Danser AHJ, Fauser BCJM, Laven JSE, Wermer MJH, Terwindt GM, Maassen Van Den Brink A. Decreased role of neuropeptides in the microvascular function in migraine patients with polycystic ovary syndrome. Atherosclerosis 2023; 384:117172. [PMID: 37400308 DOI: 10.1016/j.atherosclerosis.2023.06.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND AND AIMS To understand pathophysiological mechanisms underlying migraine as a cardiovascular risk factor, we studied neuropeptide action and endothelial function as measures of peripheral microvascular function in middle-aged women with or without migraine. METHODS We included women with the endocrine disorder polycystic ovary syndrome (PCOS), a population with supposed elevated cardiovascular risk, with and without comorbid migraine. In 26 women without and 23 women with migraine in the interictal phase (mean age 50.8 ± 2.9 years) local thermal hyperemia (LTH) of the skin of the volar forearm was measured cross-sectionally under control conditions, after inhibition of neuropeptide release by 5% lidocaine/prilocaine (EMLA) cream application, and after inhibition of nitric oxide formation by iontophoresis of NG-monomethyl-l-arginine (L-NMMA). Hereafter, changes in the natural logarithm of the reactive hyperemia index (lnRHI) and augmentation index (AI) during reperfusion after occlusion-derived ischemia were measured. RESULTS While mean values under control conditions and L-NMMA conditions were similar, migraine patients had a significantly higher mean area of the curve (AUC) of the total LTH response after EMLA application than those without (86.7 ± 26.5% versus 67.9 ± 24.2%; p = 0.014). This was also reflected by a higher median AUC of the plateau phase under similar conditions in women with migraine compared to those without (83.2% (IQR[73.2-109.5]) versus 73.2% (IQR[54.3-92.0]); p = 0.039). Mean changes in lnRHI and AI scores were similar in both groups. CONCLUSIONS In PCOS patients with migraine, neuropeptide action was lower compared with those without migraine. While larger studies are warranted, these findings provide a potential mechanism supporting previous findings that migraine may be independent from traditional risk factors, including atherosclerosis.
Collapse
Affiliation(s)
- Linda Al-Hassany
- Erasmus MC University Medical Center, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, the Netherlands
| | - Katie M Linstra
- Erasmus MC University Medical Center, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, the Netherlands; Leiden University Medical Center, Department of Neurology, Leiden, the Netherlands
| | - Cindy Meun
- Erasmus MC University Medical Center, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Rotterdam, the Netherlands
| | - Jeffrey van den Berg
- Erasmus MC University Medical Center, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, the Netherlands
| | - Eric Boersma
- Erasmus MC University Medical Center Rotterdam, Department of Cardiology, the Netherlands
| | - A H Jan Danser
- Erasmus MC University Medical Center, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, the Netherlands
| | - Bart C J M Fauser
- University Medical Center Utrecht, Department of Reproductive Medicine & Gynaecology, Utrecht, the Netherlands
| | - Joop S E Laven
- Erasmus MC University Medical Center, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Rotterdam, the Netherlands
| | - Marieke J H Wermer
- Leiden University Medical Center, Department of Neurology, Leiden, the Netherlands
| | - Gisela M Terwindt
- Leiden University Medical Center, Department of Neurology, Leiden, the Netherlands
| | - Antoinette Maassen Van Den Brink
- Erasmus MC University Medical Center, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, the Netherlands.
| |
Collapse
|
16
|
Zila-Velasque JP, Soriano-Moreno DR, Medina-Ramirez SA, Ccami-Bernal F, Castro-Diaz SD, Cortez-Soto AG, Esparza Varas AL, Fernandez-Morales J, Olortegui-Rodriguez JJ, Pelayo-Luis IP, Zafra-Tanaka JH. Prevalence of hypertension in adults living at altitude in Latin America and the Caribbean: A systematic review and meta-analysis. PLoS One 2023; 18:e0292111. [PMID: 37824544 PMCID: PMC10569637 DOI: 10.1371/journal.pone.0292111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023] Open
Abstract
OBJECTIVE The objective of this systematic review and meta-analysis was to assess the prevalence of hypertension in populations living at altitude in Latin America and the Caribbean. METHODS We conducted a systematic search from January 1, 2000 to January 10, 2023 in Web of Science (WoS)/Core Collection, WoS/Medline, WoS/Scielo, Scopus, PubMed and Embase databases. We included studies that assessed the prevalence of hypertension in altitude populations (>1500 m.a.s.l.) and these were meta-analyzed using a random-effects model. To assess the sources of heterogeneity, we performed subgroup and meta-regression analyses. RESULTS Thirty cross-sectional studies (117 406 participants) met the inclusion criteria. Studies used different cut-off points. The prevalence of hypertension in the studies that considered the cut-off point of ≥ 140/90 mmHg in the general population was 19.1%, ≥ 130/85 mmHg was 13.1%, and ≥ 130/80 mmHg was 43.4%. There was a tendency for the prevalence of hypertension to be higher in men. In meta-regression analyses, no association was found between altitude, mean age, year of publication, risk of bias and prevalence of hypertension. CONCLUSION The prevalence of hypertension in the altitude population of Latin America and the Caribbean is lower than that reported in populations living at sea level and lower than other altitude populations such as Tibetans. PROSPERO CRD42021275229.
Collapse
Affiliation(s)
- J. Pierre Zila-Velasque
- Facultad de Medicina Humana, Universidad Nacional Daniel Alcides Carrion, Pasco, Peru
- Red Latinoamericana de Medicina en Altitud e Investigacion (REDLAMAI), Pasco, Peru
| | - David R. Soriano-Moreno
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | - Sebastian A. Medina-Ramirez
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | - Fabricio Ccami-Bernal
- Facultad de Medicina, Universidad Nacional de San Agustin de Arequipa, Arequipa, Peru
| | - Sharong D. Castro-Diaz
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | - Andrea G. Cortez-Soto
- Sociedad Científica de Estudiantes de Medicina de Ica, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - Analis L. Esparza Varas
- Universidad Nacional de Trujillo, La Libertad, Peru
- Sociedad científica de estudiantes de medicina de la Universidad Nacional de Trujillo, Trujillo, Peru
| | - Jared Fernandez-Morales
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | - Juan J. Olortegui-Rodriguez
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
| | - Isabel P. Pelayo-Luis
- Unidad de Investigación Clínica y Epidemiológica, Escuela de Medicina, Universidad Peruana Unión, Lima, Peru
- Escuela de Enfermería, Universidad Peruana Unión, Lima, Peru
| | | |
Collapse
|
17
|
Kvandova M, Puzserova A, Balis P. Sexual Dimorphism in Cardiometabolic Diseases: The Role of AMPK. Int J Mol Sci 2023; 24:11986. [PMID: 37569362 PMCID: PMC10418890 DOI: 10.3390/ijms241511986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and disability among both males and females. The risk of cardiovascular diseases is heightened by the presence of a risk factor cluster of metabolic syndrome, covering obesity and obesity-related cardiometabolic risk factors such as hypertension, glucose, and lipid metabolism dysregulation primarily. Sex hormones contribute to metabolic regulation and make women and men susceptible to obesity development in a different manner, which necessitates sex-specific management. Identifying crucial factors that protect the cardiovascular system is essential to enhance primary and secondary prevention of cardiovascular diseases and should be explicitly studied from the perspective of sex differences. It seems that AMP-dependent protein kinase (AMPK) may be such a factor since it has the protective role of AMPK in the cardiovascular system, has anti-diabetic properties, and is regulated by sex hormones. Those findings highlight the potential cardiometabolic benefits of AMPK, making it an essential factor to consider. Here, we review information about the cross-talk between AMPK and sex hormones as a critical point in cardiometabolic disease development and progression and a target for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Miroslava Kvandova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (A.P.); (P.B.)
| | | | | |
Collapse
|
18
|
Sansone A, Frangione V, Lanzarotti A, Cocci A, Ceruti C, De Sio M, Imbimbo C, Mirone V, Schips L, Terrone C, Jannini EA. Effect of the new 75-mg orodispersible film of sildenafil on erection and sexual quality of life: insights from an observational study. Sex Med 2023; 11:qfac007. [PMID: 36910700 PMCID: PMC9978595 DOI: 10.1093/sexmed/qfac007] [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: 08/12/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 03/06/2023] Open
Abstract
Background The newly devised orodispersible film (ODF) of sildenafil is the first phosphodiesterase type 5 inhibitor (PDE5i) available in a 75-mg dose. This intermediate dose and the particular properties of the ODF formulation can improve the clinical management of erectile dysfunction (ED) patients. Aim We investigated the effects of the sildenafil ODF 75-mg dose on both sexual quality of life and erectile function based on the results from an observational study in daily practice in Italy. Methods This study was a post hoc analysis of results from an observational, real-life study carried out in ED patients at 6 treatment centers in Italy. All subjects were asked to take the prescribed dose of sildenafil ODF at inclusion (visit 1) and to return for a control visit (visit 2) to confirm or adapt the prescribed dose after a minimum of 4 weeks. An end of study control visit (visit 3) was performed after additional 4 weeks. Outcomes Erectile function, assessed by the International Index of Erectile Function-Erectile Function (IIEF-EF) domain; sexual quality of life, measured using the sexual quality of life instrument for men (SQoL-M). Results Among the 36 subjects initially recruited for the 75-mg dose, 5 patients dropped out of the study (2 at visit 2 and 3 at visit 3), none of whom due to treatment inefficacy or serious adverse events. At visit 2, the mean (SD) IIEF-EF scores significantly increased (∆ = 7.97 [4.71], P < 0.0001) as SQoL-M scores also did (∆ = 10.76 [10.46], P < 0.0001). At visit 3, IIEF-EF and SQoL-M scores were still significantly improved compared to baseline (∆ = 10.64 [7.01], P < 0.0001, and ∆ = 18.15 [12.32], P < 0.0001, respectively). By ANCOVA, we found no significant effects for age, BMI, previous use of PDE5i, presence of metabolic comorbidities, or smoking habits on study outcomes at both visits 2 and 3. Clinical implication The new 75-mg ODF sildenafil formulation is a safe and effective treatment for ED, significantly improving both erectile function and sexual quality of life in patients undergoing treatment. Strengths and limitations This is the first study assessing the efficacy of the sildenafil ODF 75-mg dose in a real-life setting. However, the small sample size, possible underlying cultural factors, and limited availability of clinically relevant data may have affected the reliability of our results. Conclusion The use of the 75 mg ODF formulation for sildenafil represents an effective and safe novel treatment option for ED patients.
Collapse
Affiliation(s)
- Andrea Sansone
- Section of Endocrinology and Medical Sexology (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| | | | | | - Andrea Cocci
- Section of Urology, Careggi Hospital, University of Florence IBSA Institut Biochimique SA, Pambio-Noranco, Florence, Italy
| | - Carlo Ceruti
- Division of Urology, University of Turin, Turin, Italy
| | - Marco De Sio
- Urology Unit, University "Luigi Vanvitalli" of Naples, Naples, Italy
| | - Ciro Imbimbo
- Andrology Unit, University "Federico II" of Naples, Naples, Italy
| | - Vincenzo Mirone
- Urology Unit, University "Federico II" of Naples, Naples, Italy
| | - Luigi Schips
- Department of Urology, SS Annunziata Hospital, University of Chieti, Chieti, Italy
| | - Carlo Terrone
- Department of Urology, IRCCS Ospedale Policlinico San Martino, University of Genova., Genova, Italy
| | - Emmanuele A Jannini
- Section of Endocrinology and Medical Sexology (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
19
|
Blaes AH, Nair C, Everson-Rose S, Jewett P, Wolf J, Zordoky B. Psychological measures of stress and biomarkers of inflammation, aging, and endothelial dysfunction in breast cancer survivors on aromatase inhibitors. Sci Rep 2023; 13:1677. [PMID: 36717689 PMCID: PMC9886974 DOI: 10.1038/s41598-023-28895-4] [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: 07/10/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The use of aromatase inhibitors (AIs) is associated with higher rates of cardiovascular events and lower endothelial function in breast cancer survivors. Psychosocial stress is associated with higher levels of inflammatory and aging markers, and lower endothelial function in otherwise healthy subjects. These associations among breast cancer survivors on AIs are not well defined. A cross-sectional study of 30 breast cancer survivors on AIs was performed to assess the associations between self-reported scores of psychosocial measures of depression, anxiety, and stress assessed by validated questionnaires with markers of inflammation (CRP; IL-6; IL-18), aging (p16INK4a), and endothelial function (ICAM-1, EndoPAT ratio). Significant positive correlations were observed between psychosocial measures and inflammatory markers including CRP, IL-6, and ICAM-1. However, no psychosocial scores were related to endothelial function or gene expression of the aging biomarker p16INK4a. Overall, survivors had endothelial dysfunction with reduced EndoPAT ratios. Psychosocial stress is associated with greater inflammation in breast cancer survivors on AIs, corroborating previous studies in cancer-free populations. The lack of association between psychosocial stress and either endothelial function or aging biomarkers could be due to the already low endothelial function and accelerated aging in our cohort of breast cancer survivors on AIs, though our small sample size limits conclusions. Further work in a larger and more diverse cohort of patients is needed to further understand the relationships among inflammation, aging and endothelial function in breast cancer survivors.
Collapse
Affiliation(s)
- Anne H Blaes
- Department of Medicine, University of Minnesota, Minneapolis, USA. .,Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, USA.
| | - Chandini Nair
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, USA
| | | | - Patricia Jewett
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Jack Wolf
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - Beshay Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, USA
| |
Collapse
|
20
|
Worley ML, Reed EL, Chapman CL, Kueck P, Seymour L, Fitts T, Zazulak H, Schlader ZJ, Johnson BD. Acute beetroot juice consumption does not alter cerebral autoregulation or cardiovagal baroreflex sensitivity during lower-body negative pressure in healthy adults. Front Hum Neurosci 2023; 17:1115355. [PMID: 36742355 PMCID: PMC9892911 DOI: 10.3389/fnhum.2023.1115355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Introduction Beetroot juice (BRJ) improves peripheral endothelial function and vascular compliance, likely due to increased nitric oxide bioavailability. It is unknown if BRJ alters cerebrovascular function and cardiovagal baroreflex control in healthy individuals. Purpose We tested the hypotheses that BRJ consumption improves cerebral autoregulation (CA) and cardiovagal baroreflex sensitivity (cBRS) during lower-body negative pressure (LBNP). Methods Thirteen healthy adults (age: 26 ± 4 years; 5 women) performed oscillatory (O-LBNP) and static LBNP (S-LBNP) before (PRE) and 3 h after consuming 500 mL of BRJ (POST). Participants inhaled 3% CO2 (21% O2, 76% N2) during a 5 min baseline and throughout LBNP to attenuate reductions in end-tidal CO2 tension (PETCO2). O-LBNP was conducted at ∼0.02 Hz for six cycles (-70 mmHg), followed by a 3-min recovery before S-LBNP (-40 mmHg) for 7 min. Beat-to-beat middle cerebral artery blood velocity (MCAv) (transcranial Doppler) and blood pressure were continuously recorded. CA was assessed using transfer function analysis to calculate coherence, gain, and phase in the very-low-frequency (VLF; 0.020-0.070 Hz) and low-frequency bands (LF; 0.07-0.20 Hz). cBRS was calculated using the sequence method. Comparisons between POST vs. PRE are reported as mean ± SD. Results During O-LBNP, coherence VLF was greater at POST (0.55 ± 0.06 vs. 0.46 ± 0.08; P < 0.01), but phase VLF (P = 0.17) and gain VLF (P = 0.69) were not different. Coherence LF and phase LF were not different, but gain LF was lower at POST (1.03 ± 0.20 vs. 1.12 ± 0.30 cm/s/mmHg; P = 0.05). During S-LBNP, CA was not different in the VLF or LF bands (all P > 0.10). Up-cBRS and Down-cBRS were not different during both LBNP protocols. Conclusion These preliminary data indicate that CA and cBRS during LBNP in healthy, young adults is largely unaffected by an acute bolus of BRJ.
Collapse
Affiliation(s)
- Morgan L. Worley
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Emma L. Reed
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Human Physiology, Bowerman Sports Science Center, University of Oregon, Eugene, OR, United States
| | - Christopher L. Chapman
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Human Physiology, Bowerman Sports Science Center, University of Oregon, Eugene, OR, United States
| | - Paul Kueck
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Lauren Seymour
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Triniti Fitts
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Hannah Zazulak
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Zachary J. Schlader
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Kinesiology, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| | - Blair D. Johnson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Kinesiology, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
21
|
Tsirka A. Gender Differences in Cardiovascular Health: Hormonal Effects on Cardiovascular Risk and Management. Handb Exp Pharmacol 2023; 282:201-218. [PMID: 37468722 DOI: 10.1007/164_2023_668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Cardiovascular disease is the number one cause of death worldwide both in men and women. Significant differences however exist in the age of onset, type of cardiovascular disease (CVD), and treatment response among the genders. These differences are due to both gender societal effects and biological differences from the influence of the hormonal and metabolic milieu in the two sexes.Estrogens are the main female hormone. They impact signaling in almost every body organ and thus affect cardiovascular risk factors. These result in differential expression of CVD. The differential effect of treatment modalities is only recently being evaluated.Research is ongoing regarding the pathophysiology of these differences. Our current level of knowledge is described in this chapter.
Collapse
Affiliation(s)
- Anna Tsirka
- University of Connecticut School of Medicine, Farmington, CT, USA.
- Connecticut Children's Medical Center, Hartford, CT, USA.
- St Christopher's Children's Hospital, Drexel University Medical School, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Hyperuricemia and Endothelial Function: Is It a Simple Association or Do Gender Differences Play a Role in This Binomial? Biomedicines 2022; 10:biomedicines10123067. [PMID: 36551823 PMCID: PMC9775568 DOI: 10.3390/biomedicines10123067] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022] Open
Abstract
The endothelium plays a fundamental role in the biological processes that ensure physiological vessel integrity, synthesizing numerous substances that are capable of modulating the tone of vessels, inflammation and the immune system, and platelet function. Endothelial dysfunction refers to an anomaly that develops at the level of the tunica that lines the internal surface of arterial and venous vessels, or, more precisely, an alteration to normal endothelial function, which involves the loss of some structural and/or functional characteristics. Studies on sex differences in endothelial function are conflicting, with some showing an earlier decline in endothelial function in men compared to women, while others show a similar age of onset between the sexes. Since increased cardiovascular risk coincides with menopause, female hormones, particularly estrogen, are generally believed to be cardioprotective. Furthermore, it is often proposed that androgens are harmful. In truth, these relationships are more complex than one might think and are not just dependent on fluctuations in circulating hormones. An increase in serum uric acid is widely regarded as a possible risk factor for cardiovascular disease; however, its role in the occurrence of endothelial dysfunction has not yet been elucidated. Several studies in the literature have evaluated sex-related differences in the association between elevated uric acid levels and cardiovascular events, with conflicting results. The association between uric acid and cardiovascular disease is still controversial, and it is not yet clear how gender differences affect the serum concentration of these substances. This review was primarily aimed at clarifying the effects of uric acid at the level of the vascular endothelium and describing how it could theoretically cause damage to endothelial integrity. The second aim was to determine if there are gender differences in uric acid metabolism and how these differences interact with the vascular endothelium.
Collapse
|
23
|
Miyata I, Nagai A. Distinctive nail pattern following androgen replacement therapy. IJU Case Rep 2022; 5:515-516. [PMID: 36341204 PMCID: PMC9626338 DOI: 10.1002/iju5.12530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/17/2022] [Indexed: 11/12/2022] Open
Abstract
Introduction A distinctive nail pattern in a patient undergoing androgen replacement therapy is presented hereby. Case presentation A 47‐year‐old male patient noticed a peculiar “washboard‐like” pattern on his fingernails. He had been undergoing androgen replacement therapy for late onset hypogonadism syndrome. The patient realized his nails were growing faster after his therapy was altered from a tri‐weekly basis to a bi‐weekly basis. Conclusion This phenomenon is likely to be attributed to the androgen replacement therapy, as the spacing between the ridges were widening in concordance with the strengthening of the therapy.
Collapse
Affiliation(s)
- Ippei Miyata
- Department of Pediatrics Kawasaki Medical School Okayama Japan
| | | |
Collapse
|
24
|
Somani YB, Soares RN, Gosalia J, Delgado JM, Flanagan M, Basu S, Kim-Shapiro DB, Murias JM, Proctor DN. A single dose of dietary nitrate supplementation protects against endothelial ischemia-reperfusion injury in early postmenopausal women. Appl Physiol Nutr Metab 2022; 47:749-761. [PMID: 35358395 PMCID: PMC10941101 DOI: 10.1139/apnm-2021-0693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The onset of menopause and accompanying changes to ovarian hormones often precedes endothelial dysfunction in women. In particular, accelerated impairments in macrovascular and microvascular function coincide with the loss of estrogen, as does impaired endothelial responses to ischemia-reperfusion (IR) injury. In healthy, early postmenopausal women (n = 12; 3.9 ± 1.5 years since menopause) we tested the hypothesis that acute dietary nitrate (NO3-) supplementation would improve endothelial function and attenuate the magnitude of endothelial dysfunction following whole-arm IR in comparison with placebo. In this randomized, double-blind, placebo-controlled, crossover study we tested participants before and after NO3--rich (BRnitrate) and NO3--depleted (BRplacebo) beetroot juice (BR) consumption, as well as following IR injury, and 15 min after IR to assess recovery. Analyses with repeated-measures general linear models revealed a condition × time interaction for brachial artery flow-mediated dilation (FMD; P = 0.04), and no interaction effect was found for the near-infrared spectroscopy-derived reperfusion slope (P = 0.86). Follow-up analysis showed a significant decline in FMD following IR injury with BRplacebo in comparison with all other timepoints (all, P < 0.05), while this decline was not present with BRnitrate (all, P > 0.05). Our findings demonstrate that a single dose of dietary NO3- minimizes IR-induced macrovascular endothelial dysfunction in healthy, early postmenopausal women, but does not improve resting macrovascular and microvascular function. Trial registration number: NCT03644472. Novelty: In healthy, early postmenopausal women, a single dose of NO3--rich BR can protect against IR-induced endothelial dysfunction. This protection may be due to nitric oxide bioactivity during IR rather than improved endothelial function prior to the IR protocol per se.
Collapse
Affiliation(s)
- Y B Somani
- Department of Kinesiology, Integrative Vascular Physiology Lab, Penn State University, University Park, PA, USA
| | - R N Soares
- Dalton Cardiovascular Research Center, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - J Gosalia
- Department of Kinesiology, Integrative Vascular Physiology Lab, Penn State University, University Park, PA, USA
| | - J M Delgado
- Department of Kinesiology, Integrative Vascular Physiology Lab, Penn State University, University Park, PA, USA
| | - M Flanagan
- Penn State Hershey Family and Community Medicine, University Park, PA, USA
| | - S Basu
- Translational Science Center and Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - D B Kim-Shapiro
- Translational Science Center and Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - J M Murias
- Department of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - D N Proctor
- Department of Kinesiology, Integrative Vascular Physiology Lab, Penn State University, University Park, PA, USA
| |
Collapse
|
25
|
Mizrak I, Asserhøj LL, Lund MAV, Greisen GO, Clausen TD, Main KM, Vejlstrup NG, Jensen RB, Pinborg A, Madsen PL. Aortic distensibility is equal in prepubertal girls and boys and increases with puberty in girls. Am J Physiol Heart Circ Physiol 2022; 323:H312-H321. [PMID: 35687504 DOI: 10.1152/ajpheart.00097.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Windkessel function is governed by conductance artery compliance that is associated with cardiovascular disease in adults independently of other risk factors. Sex-related differences in conductance artery compliance partly explain the sex-related differences in risk of cardiovascular disease. Studies on sex-related differences in conductance artery function in prepubertal children are few and inconclusive. This study determined conductance artery compliance and cardiac function by magnetic resonance imaging in 150 healthy children (75 girls) aged 7-10 years. Any sex-related difference in conductance artery function was determined with correction for other potential predictors in multivariable linear regression models. Our data showed that ascending (crude mean difference 1.11 95% CI (0.22; 2.01)) and descending (crude mean difference 1.10 95% CI (0.09; 1.91)) aortic distensibility were higher in girls, but differences disappeared after adjustment for pubertal status and other identified potential predictors. Systolic and diastolic blood pressure, cardiac output, left ventricle (LV) systolic function, and total peripheral resistance did not differ between the sexes. In girls, heart rate was 7 bpm higher, whereas pulse pressure (by 2 mmHg), and LV end-diastolic volume index (by 7 mL) and stroke volume (by 5 mL) were lower. LV peak filling rate indexed to LV end-diastolic volume was 0.5 s-1 higher in girls. In conclusion, prepubertal girls and boys have equal conductance artery function. Thus, the well-known sex difference in adult conductance artery function seems to develop after the onset of puberty with girls initially increasing aortic distensibility.
Collapse
Affiliation(s)
- Ikram Mizrak
- Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Fertility Department, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Louise L Asserhøj
- Fertility Department, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Growth and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten A V Lund
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gorm O Greisen
- Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tine D Clausen
- Department of Gynecology and Obstetrics, North Zealand Hospital, Copenhagen University Hospital, Hilleroed, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Niels G Vejlstrup
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rikke B Jensen
- Department of Growth and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anja Pinborg
- Fertility Department, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Per L Madsen
- Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Inoue K, Fujie S, Horii N, Yamazaki H, Uchida M, Iemitsu M. Aerobic exercise training-induced follistatin-like 1 secretion in the skeletal muscle is related to arterial stiffness via arterial NO production in obese rats. Physiol Rep 2022; 10:e15300. [PMID: 35585770 PMCID: PMC9117810 DOI: 10.14814/phy2.15300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 11/25/2022] Open
Abstract
Follistatin‐like 1 (FSTL1), which is mainly secreted from skeletal muscle and myocardium, upregulates protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) phosphorylation in vascular endothelial cells. It is unclear whether skeletal muscle‐ and myocardium‐derived FSTL1 secretion induced by aerobic exercise training is involved in the reduction of arterial stiffness via arterial NO production in obese rats. This study aimed to clarify whether aerobic exercise training‐induced FSTL1 secretion in myocardium and skeletal muscle is associated with a reduction in arterial stiffness via arterial Akt‐eNOS signaling pathway in obese rats. Sixteen Otsuka Long‐Evans Tokushima Fatty (OLETF) obese rats were randomly divided into two groups: sedentary control (OLETF‐CON) and eight‐week aerobic exercise training (treadmill for 60min at 25m/min, 5days/week, OLETF‐AT). Eight Long‐Evans Tokushima Otsuka (LETO) rats were used as a healthy sedentary control group. In OLETF‐CON, serum FSTL1, arterial Akt and eNOS phosphorylation, and arterial nitrite/nitrate (NOx) levels were significantly lower, and carotid‐femoral pulse wave velocity (cfPWV) was significantly greater than those in LETO. These parameters were improved in the OLETF‐AT compared to the OLETF‐CON. In the OLETF‐AT, FSTL1 levels in slow‐twitch fiber‐rich soleus muscle were significantly greater than those in the OLETF‐CON, but not in myocardium, fast‐twitch fiber‐rich tibialis anterior muscle, and adipose tissue. Serum FSTL1 levels were positively correlated with soleus FSTL1, arterial eNOS phosphorylation, and NOx levels and negatively correlated with cfPWV. Thus, aerobic exercise training‐induced FSTL1 secretion in slow‐twitch fiber‐rich muscles may be associated with a reduction in arterial stiffness via arterial NO production in obese rats.
Collapse
Affiliation(s)
- Kenichiro Inoue
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Shumpei Fujie
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Naoki Horii
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Henry Yamazaki
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Masataka Uchida
- Global Innovation Research Organization, Ritsumeikan University, Shiga, Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| |
Collapse
|
27
|
Gajecki D, Gawryś J, Szahidewicz-Krupska E, Doroszko A. Role of Erythrocytes in Nitric Oxide Metabolism and Paracrine Regulation of Endothelial Function. Antioxidants (Basel) 2022; 11:antiox11050943. [PMID: 35624807 PMCID: PMC9137828 DOI: 10.3390/antiox11050943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 01/27/2023] Open
Abstract
Emerging studies provide new data shedding some light on the complex and pivotal role of red blood cells (RBCs) in nitric oxide (NO) metabolism and paracrine regulation of endothelial function. NO is involved in the regulation of vasodilatation, platelet aggregation, inflammation, hypoxic adaptation, and oxidative stress. Even though tremendous knowledge about NO metabolism has been collected, the exact RBCs’ status still requires evaluation. This paper summarizes the actual knowledge regarding the role of erythrocytes as a mobile depot of amino acids necessary for NO biotransformation. Moreover, the complex regulation of RBCs’ translocases is presented with a particular focus on cationic amino acid transporters (CATs) responsible for the NO substrates and derivatives transport. The main part demonstrates the intraerythrocytic metabolism of L-arginine with its regulation by reactive oxygen species and arginase activity. Additionally, the process of nitrite and nitrate turnover was demonstrated to be another stable source of NO, with its reduction by xanthine oxidoreductase or hemoglobin. Additional function of hemoglobin in NO synthesis and its subsequent stabilization in steady intermediates is also discussed. Furthermore, RBCs regulate the vascular tone by releasing ATP, inducing smooth muscle cell relaxation, and decreasing platelet aggregation. Erythrocytes and intraerythrocytic NO metabolism are also responsible for the maintenance of normotension. Hence, RBCs became a promising new therapeutic target in restoring NO homeostasis in cardiovascular disorders.
Collapse
|
28
|
Jannini TB, Sansone A, Rossi R, Di Lorenzo G, Toscano M, Siracusano A, Jannini EA. Pharmacological strategies for sexual recovery in men undergoing antipsychotic treatment. Expert Opin Pharmacother 2022; 23:1065-1080. [PMID: 35470768 DOI: 10.1080/14656566.2022.2071124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION : First- and second-generation antipsychotics are highly accountable for causing a plethora of medical side effects, ranging from metabolic imbalances to sexual dysfunction (SD), that frequently undermine patient-doctor relationships. Nevertheless, to date antipsychotics are one of the best treatment options for dealing with numerous either acute or chronic conditions like agitation, suicidality, depression, dementia, and of course psychosis. For these reasons, clinicians need to handle them wisely to preserve patients' sexual health, avoid poor therapeutic adherence and prevent high rates of therapy drop-out. AREAS COVERED : This article reviews the literature on pharmacologic approaches for management strategies in men who are administered with antipsychotics and developed SD. The etiology of antipsychotic-induced SD is also discussed. EXPERT OPINION : Clinicians must consider sexual life as a major health domain. To do so, a first step would be to measure and monitor sexual function by means of psychometric tools. Secondly, primary prevention should be conducted when choosing antipsychotics, i.e., picking sex-sparing compounds like aripiprazole or brexpiprazole. Thirdly, if sexolytic compounds cannot be dismissed, such as first-generation antipsychotics, risperidone, paliperidone, or amisulpride, then aripiprazole 5-20 mg/day adjunctive therapy has proven to be most effective in normalizing prolactin levels and consequently treating antipsychotic-induced SD.
Collapse
Affiliation(s)
- Tommaso B Jannini
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Sansone
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Rodolfo Rossi
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giorgio Di Lorenzo
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS-Fondazione Santa Lucia, Rome, Italy
| | - Massimiliano Toscano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy.,Department of Neurology, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Alberto Siracusano
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emmanuele A Jannini
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
29
|
Qiu Q, Zhou X, Wu L, Zhang Y, Yu Z, Wang M, Huang H, Luo X, Pan D. Serum Cortisol Is Associated With Cerebral Small Vessel Disease-Related Brain Changes and Cognitive Impairment. Front Aging Neurosci 2022; 13:809684. [PMID: 35126094 PMCID: PMC8814515 DOI: 10.3389/fnagi.2021.809684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022] Open
Abstract
Objective To evaluate the relationship between serum cortisol, cerebral small vessel disease (CSVD) neuroimaging markers, and cognitive performance. Methods We recruited patients over 50 years old who attended our hospital for physical examination between November 2020 and July 2021. All participants were subject to brain magnetic resonance imaging (MRI), serum cortisol examination, and the Montreal cognitive function assessment (MoCA). On brain MRI, we scored the presence of each marker of CSVD, including white matter hyperintensity (WMH), lacunes, cerebral microbleeds (CMBs), and enlarged perivascular spaces (EPVS). One point was awarded for the presence of each marker, producing a score between 0 and 4. Results In total, 158 participants were included in this study with a mean age of 60.5 (56.0–66.3) years; 55.1% were male. In the multivariable analyses, serum cortisol level was an independent predictor of WMH severity, the presence of lacunes/CMBs, moderate-severe EPVS and total CSVD burden after adjusting for confounding factors. Serum cortisol level had positive associations with periventricular/deep Fazekas score, burdens of lacunes/CMBs, moderate-severe EPVS, and total CSVD burden in dose-dependent manner, and was an independent predictor of cognitive impairment. Furthermore, the results of the receiver operating characteristic (ROC) curve analysis revealed an area under curve (AUC) of 0.745 with 64.1% sensitivity and 82.5% specificity, and an AUC of 0.705 with 52.1% sensitivity and 85.5 specificity of cortisol in detecting patients with high CSVD burden and MCI, respectively. Conclusions Serum cortisol level is independently associated with each CSVD MRI markers, total CSVD burden and cognitive impairment. These findings provide clues for pathological mechanisms and suggest serum cortisol as a promising biomarker associated with CSVD.
Collapse
|
30
|
Lin Z, Ding Q, Li X, Feng Y, He H, Huang C, Zhu Y. Targeting Epigenetic Mechanisms in Vascular Aging. Front Cardiovasc Med 2022; 8:806988. [PMID: 35059451 PMCID: PMC8764463 DOI: 10.3389/fcvm.2021.806988] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/30/2021] [Indexed: 12/28/2022] Open
Abstract
Environment, diseases, lack of exercise, and aged tendency of population have becoming crucial factors that induce vascular aging. Vascular aging is unmodifiable risk factor for diseases like diabetes, hypertension, atherosclerosis, and hyperlipidemia. Effective interventions to combat this vascular function decline is becoming increasingly urgent as the rising hospitalization rate caused by vascular aging-related diseases. Fortunately, recent transformative omics approaches have enabled us to examine vascular aging mechanisms at unprecedented levels and precision, which make our understanding of slowing down or reversing vascular aging become possible. Epigenetic viz. DNA methylation, histone modifications, and non-coding RNA-based mechanisms, is a hallmark of vascular aging, its deregulation leads to aberrant transcription changes in tissues. Epigenetics mechanisms by mediating covalent modifications to DNA and histone proteins, consequently, influence the sensitivity and activities of signaling pathways in cells and tissues. A growing body of evidence supports correlations between epigenetic changes and vascular aging. In this article, we will provide a comprehensive overview of epigenetic changes associated with vascular aging based on the recent findings with a focus on molecular mechanisms of action, strategies to reverse epigenetic changes, and future perspectives.
Collapse
Affiliation(s)
- Zhongxiao Lin
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
- Key Laboratory of Molecular Target and Clinical Pharmacology and National Key Laboratory of Respiratory Diseases, School of Pharmaceutic Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - Xinzhi Li
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - Yuliang Feng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Hao He
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - Chuoji Huang
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - YiZhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Odom MR, Pak ES, Hannan JL. Ex vivo Akt inhibition reverses castration induced internal pudendal artery and penile endothelial dysfunction. Life Sci 2021; 285:119966. [PMID: 34543641 DOI: 10.1016/j.lfs.2021.119966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/25/2022]
Abstract
AIMS Androgen deprivation therapy is a common prostate cancer treatment which causes men to have castrate levels of testosterone. Unfortunately, most testosterone deficient patients will suffer severe erectile dysfunction (ED) and have no effective ED treatment options. Testosterone deficiency causes endothelial dysfunction and impairs penile vasodilation necessary to maintain an erection. Recent evidence demonstrates testosterone activates androgen receptors (AR) and generates nitric oxide (NO) through the Akt-endothelial NO synthase (eNOS) pathway; however, it remains unknown how castration impacts this signaling pathway. MATERIALS AND METHODS In this study, we used a surgically castrated rat model to determine how castration impacts ex vivo internal pudendal artery (IPA) and penile relaxation through the Akt-eNOS pathway. KEY FINDINGS Unlike systemic vasculature, castration causes significant IPA and penis endothelial dysfunction associated with a 50% AR reduction. Though testosterone and acetylcholine (ACh) both phosphorylate Akt and eNOS, castration did not affect testosterone-mediated IPA and penile Akt or eNOS phosphorylation. Surprisingly, castration increases ACh-mediated Akt and eNOS phosphorylation but reduces the eNOS dimer to monomer ratio. Akt inhibition using 10DEBC preserves IPA eNOS dimers. Functionally, 10DEBC reverses castration induced ex vivo IPA and penile endothelial dysfunction. SIGNIFICANCE These data demonstrate how castration uncouples eNOS and provide a novel strategy for improving endothelial-dependent relaxation necessary for an erection. Further studies are needed to determine if Akt inhibition may treat or even prevent ED in testosterone deficient prostate cancer survivors.
Collapse
Affiliation(s)
- Michael R Odom
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA; Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Elena S Pak
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Johanna L Hannan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
32
|
Baranauskas MN, Freemas JA, Tan R, Carter SJ. Moving beyond inclusion: Methodological considerations for the menstrual cycle and menopause in research evaluating effects of dietary nitrate on vascular function. Nitric Oxide 2021; 118:39-48. [PMID: 34774755 DOI: 10.1016/j.niox.2021.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/18/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
Recent reports have acknowledged the underrepresentation of women in the field of dietary nitrate (NO3-) research. Undoubtedly, greater participation from women is warranted to clarify potential sex differences in the responses to dietary NO3- interventions. However, careful consideration for the effects of sex hormones - principally 17β-estradiol - on endogenous nitric oxide (NO) synthesis and dietary NO3- reductase capacity is necessary for improved interpretation and reproducibility of such investigations. From available literature, we present a narrative review describing how hormonal variations across the menstrual cycle, as well as with menopause, may impact NO biosynthesis catalyzed by NO synthase enzymes and NO3- reduction via the enterosalivary pathway. In doing so, we address methodological considerations related to the menstrual cycle and hormonal contraceptive use relevant for the inclusion of premenopausal women along with factors to consider when testing postmenopausal women. Adherence to such methodological practices may explicate the utility of dietary NO3- supplementation as a means to improve vascular function among women across the lifespan.
Collapse
Affiliation(s)
- Marissa N Baranauskas
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA.
| | - Jessica A Freemas
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA
| | - Rachel Tan
- Department of Natural Science, Seaver College, Pepperdine University, 90263, USA
| | - Stephen J Carter
- Department of Kinesiology, School of Public Health - Bloomington, Bloomington, Indiana University, 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| |
Collapse
|
33
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2021; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C. Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R. Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
34
|
Schereider IRG, Vassallo DV, Simões MR. Chronic mercury exposure induces oxidative stress in female rats by endothelial nitric oxide synthase uncoupling and cyclooxygenase-2 activation, without affecting oestrogen receptor function. Basic Clin Pharmacol Toxicol 2021; 129:470-485. [PMID: 34491608 DOI: 10.1111/bcpt.13655] [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: 12/02/2020] [Revised: 08/04/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022]
Abstract
Mercury has been shown to be a significant health risk factor and is positively associated with cardiovascular diseases. Evidence reveals that men are more likely to develop cardiovascular diseases than women during reproductive age. However, the effects of mercury in females remain poorly investigated, despite the finding that female hormones demonstrate a cardioprotective role. In the present study, we evaluated whether chronic mercury chloride exposure could alter blood pressure and vascular function of the female rat aorta. Ten-week-old female Wistar rats were divided into two groups: control (vehicle) and mercury treated (first dose of 4.6 μg/kg, subsequent daily doses of 0.07 μg/kg), im. Mercury treatment did not modify systolic blood pressure (SBP) but increased vascular reactivity due to the reduction of nitric oxide bioavailability associated with the increase in reactive oxygen species from endothelial nitric oxide synthase (eNOS) uncoupling. Furthermore, increased participation of the cyclooxygenase-2 pathway occurred through an imbalance in thromboxane 2 and prostacyclin 2. However, the oestrogen signalling pathway was not altered in either group. These results demonstrated that chronic exposure to mercury in females induced endothelial dysfunction and, consequently, increased aortic vascular reactivity, causing vascular damage to the female rat aorta and representing a risk of cardiovascular diseases.
Collapse
Affiliation(s)
| | - Dalton Valentim Vassallo
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil.,Health Science Center of Vitória, School of Sciences of Santa Casa de Misericórdia de Vitória - EMESCAM, Vitória, Espírito Santo, Brazil
| | - Maylla Ronacher Simões
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
35
|
The role of extracellular matrix in tumour angiogenesis: the throne has NOx servants. Biochem Soc Trans 2021; 48:2539-2555. [PMID: 33150941 PMCID: PMC7752075 DOI: 10.1042/bst20200208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) dynamics in tumour tissue are deregulated compared to the ECM in healthy tissue along with disorganized architecture and irregular behaviour of the residing cells. Nitric oxide (NO) as a pleiotropic molecule exerts different effects on the components of the ECM driving or inhibiting augmented angiogenesis and tumour progression and tumour cell proliferation and metastasis. These effects rely on the concentration of NO within the tumour tissue, the nature of the surrounding microenvironment and the sensitivity of resident cells to NO. In this review article, we summarize the recent findings on the correlation between the levels of NO and the ECM components towards the modulation of tumour angiogenesis in different types of cancers. These are discussed principally in the context of how NO modulates the expression of ECM proteins resulting in either the promotion or inhibition of tumour growth via tumour angiogenesis. Furthermore, the regulatory effects of individual ECM components on the expression of the NO synthase enzymes and NO production were reviewed. These findings support the current efforts for developing effective therapeutics for cancers.
Collapse
|
36
|
Zein JG, McManus JM, Sharifi N, Erzurum SC, Marozkina N, Lahm T, Giddings O, Davis MD, DeBoer MD, Comhair SA, Bazeley P, Kim HJ, Busse W, Calhoun W, Castro M, Chung KF, Fahy JV, Israel E, Jarjour NN, Levy BD, Mauger DT, Moore WC, Ortega VE, Peters M, Bleecker ER, Meyers DA, Zhao Y, Wenzel SE, Gaston B. Benefits of Airway Androgen Receptor Expression in Human Asthma. Am J Respir Crit Care Med 2021; 204:285-293. [PMID: 33779531 DOI: 10.1164/rccm.202009-3720oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Androgens are potentially beneficial in asthma, but AR (androgen receptor) has not been studied in human airways.Objectives: To measure whether AR and its ligands are associated with human asthma outcomes.Methods: We compared the effects of AR expression on lung function, symptom scores, and fractional exhaled nitric oxide (FeNO) in adults enrolled in SARP (Severe Asthma Research Program). The impact of sex and of androgens on asthma outcomes was also evaluated in the SARP with validation studies in the Cleveland Clinic Health System and the NHANES (U.S. National Health and Nutrition Examination Survey).Measurements and Main Results: In SARP (n = 128), AR gene expression from bronchoscopic epithelial brushings was positively associated with both FEV1/FVC ratio (R2 = 0.135, P = 0.0002) and the total Asthma Quality of Life Questionnaire score (R2 = 0.056, P = 0.016) and was negatively associated with FeNO (R2 = 0.178, P = 9.8 × 10-6) and NOS2 (nitric oxide synthase gene) expression (R2 = 0.281, P = 1.2 × 10-10). In SARP (n = 1,659), the Cleveland Clinic Health System (n = 32,527), and the NHANES (n = 2,629), women had more asthma exacerbations and emergency department visits than men. The levels of the AR ligand precursor dehydroepiandrosterone sulfate correlated positively with the FEV1 in both women and men.Conclusions: Higher bronchial AR expression and higher androgen levels are associated with better lung function, fewer symptoms, and a lower FeNO in human asthma. The role of androgens should be considered in asthma management.
Collapse
Affiliation(s)
- Joe G Zein
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | - Serpil C Erzurum
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | | | - Mark D DeBoer
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Suzy A Comhair
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Peter Bazeley
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Hyun Jo Kim
- Department of Systems Biology and Bioinformatics, Case Western Reserve University, Cleveland, Ohio
| | - William Busse
- Department of Medicine, School of Medicine, University of Wisconsin, Madison, Wisconsin
| | - William Calhoun
- Department of Medicine, University of Texas Medical Branch, University of Texas, Galveston, Texas
| | - Mario Castro
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of Kansas, Kansas City, Kansas
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John V Fahy
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of Kansas, Kansas City, Kansas.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Nizar N Jarjour
- Department of Medicine, School of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - David T Mauger
- Center for Biostatistics and Epidemiology, School of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Wendy C Moore
- Section on Pulmonary, Critical Care, Allergic, and Immunologic Disease, Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Victor E Ortega
- Section on Pulmonary, Critical Care, Allergic, and Immunologic Disease, Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Michael Peters
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Eugene R Bleecker
- Division of Genetics, Genomics, and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | - Deborah A Meyers
- Division of Genetics, Genomics, and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | - Yi Zhao
- Department of Biostatistics and Health Science Data, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
37
|
Effects of sex and estrous cycle on the brain and plasma arginine metabolic profile in rats. Amino Acids 2021; 53:1441-1454. [PMID: 34245369 DOI: 10.1007/s00726-021-03040-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
L-arginine is a versatile amino acid with a number of bioactive metabolites. Increasing evidence implicates altered arginine metabolism in the aging and neurodegenerative processes. The present study, for the first time, determined the effects of sex and estrous cycle on the brain and blood (plasma) arginine metabolic profile in naïve rats. Female rats displayed significantly lower levels of L-arginine in the frontal cortex and three sub-regions of the hippocampus when compared to male rats. Moreover, female rats had significantly higher levels of L-arginine and γ-aminobutyric acid, but lower levels of L-ornithine, agmatine and putrescine, in plasma relative to male rats. The observed sex difference in brain L-arginine appeared to be independent of the enzymes involved in its metabolism, de novo synthesis and blood-to-brain transport (cationic acid transporter 1 protein expression at least), as well as circulating L-arginine. While the estrous cycle did not affect L-arginine and its metabolites in the brain, there were estrous cycle phase-dependent changes in plasma L-arginine. These findings demonstrate the sex difference in brain L-arginine in the estrous cycle-independent manner. Since peripheral blood has been increasingly used to identify biomarkers of brain pathology, the influences of sex and estrous cycle on blood arginine metabolic profile need attention when experimental research involves female rodents.
Collapse
|
38
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
39
|
Mauvais-Jarvis F, Berthold HK, Campesi I, Carrero JJ, Dakal S, Franconi F, Gouni-Berthold I, Heiman ML, Kautzky-Willer A, Klein SL, Murphy A, Regitz-Zagrosek V, Reue K, Rubin JB. Sex- and Gender-Based Pharmacological Response to Drugs. Pharmacol Rev 2021; 73:730-762. [PMID: 33653873 PMCID: PMC7938661 DOI: 10.1124/pharmrev.120.000206] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In humans, the combination of all sex-specific genetic, epigenetic, and hormonal influences of biologic sex produces different in vivo environments for male and female cells. We dissect how these influences of sex modify the pharmacokinetics and pharmacodynamics of multiple drugs and provide examples for common drugs acting on specific organ systems. We also discuss how gender of physicians and patients may influence the therapeutic response to drugs. We aim to highlight sex as a genetic modifier of the pharmacological response to drugs, which should be considered as a necessary step toward precision medicine that will benefit men and women. SIGNIFICANCE STATEMENT: This study discusses the influences of biologic sex on the pharmacokinetics and pharmacodynamics of drugs and provides examples for common drugs acting on specific organ systems. This study also discusses how gender of physicians and patients influence the therapeutic response to drugs.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Heiner K Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ilaria Campesi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Juan-Jesus Carrero
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Santosh Dakal
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Flavia Franconi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ioanna Gouni-Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Mark L Heiman
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Alexandra Kautzky-Willer
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Sabra L Klein
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Anne Murphy
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Vera Regitz-Zagrosek
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Karen Reue
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Joshua B Rubin
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| |
Collapse
|
40
|
Mintz J, Vedenko A, Rosete O, Shah K, Goldstein G, Hare JM, Ramasamy R, Arora H. Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics. Vaccines (Basel) 2021; 9:94. [PMID: 33513777 PMCID: PMC7912608 DOI: 10.3390/vaccines9020094] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a short-lived, ubiquitous signaling molecule that affects numerous critical functions in the body. There are markedly conflicting findings in the literature regarding the bimodal effects of NO in carcinogenesis and tumor progression, which has important consequences for treatment. Several preclinical and clinical studies have suggested that both pro- and antitumorigenic effects of NO depend on multiple aspects, including, but not limited to, tissue of generation, the level of production, the oxidative/reductive (redox) environment in which this radical is generated, the presence or absence of NO transduction elements, and the tumor microenvironment. Generally, there are four major categories of NO-based anticancer therapies: NO donors, phosphodiesterase inhibitors (PDE-i), soluble guanylyl cyclase (sGC) activators, and immunomodulators. Of these, NO donors are well studied, well characterized, and also the most promising. In this study, we review the current knowledge in this area, with an emphasis placed on the role of NO as an anticancer therapy and dysregulated molecular interactions during the evolution of cancer, highlighting the strategies that may aid in the targeting of cancer.
Collapse
Affiliation(s)
- Joel Mintz
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA;
| | - Anastasia Vedenko
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
| | - Omar Rosete
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Khushi Shah
- College of Arts and Sciences, University of Miami, Miami, FL 33146, USA;
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL 32816, USA;
| | - Joshua M. Hare
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Medicine, Cardiology Division, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Himanshu Arora
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
41
|
Association of waist-to-height ratio with estimated glomerular filtration rate in middle-aged and elderly Chinese. ACTA ACUST UNITED AC 2020; 28:407-412. [PMID: 33350668 DOI: 10.1097/gme.0000000000001718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study aimed to assess the relationship between waist-to-height (WHtR) and estimated glomerular filtration rate (eGFR) in men, non-menopausal, and postmenopausal women among middle-aged and elderly Chinese. METHODS This study analyzed the data of 7,807 participants in a cross-sectional survey, ie, the third wave of the China Health and Retirement Longitudinal Study. Restrictive cubic-spline regression with three knots was used to assess the dose-response association of WHtR with eGFR. Piecewise linear regression models were further established to calculate the slope of each segment and their 95% confidence interval (CI). RESULTS After adjusting for potential confounders, an inverse L-shaped dose-response relationship was found between WHtR and eGFR among men (Pnonlinear = 0.024, threshold = 0.513) and postmenopausal women (Pnonlinear = 0.009, threshold = 0.503). The slopes on the right sides of the threshold were statistically significant among men (β2 = -33.77, 95% CI: -53.23 to -14.31) and postmenopausal women (β2 = -36.53, 95% CI: -49.71 to -23.35), respectively. A weak negative linear relationship existed between WHtR and eGFR in non-menopausal women. CONCLUSIONS The relationship between WHtR and eGFR tended to be inverse-L-shaped in men and postmenopausal women, but may vary with postmenopausal status in women.
Collapse
|
42
|
Iwamoto E, Sakamoto R, Tsuchida W, Yamazaki K, Kamoda T, Neki T, Katayose M, Casey DP. Effects of menstrual cycle and menopause on internal carotid artery shear-mediated dilation in women. Am J Physiol Heart Circ Physiol 2020; 320:H679-H689. [PMID: 33306444 DOI: 10.1152/ajpheart.00810.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study aimed to elucidate the effects of change in estrogen during the menstrual cycle and menopause on shear-mediated dilation of the internal carotid artery (ICA), a potential index of cerebrovascular endothelial function. Shear-mediated dilation of the ICA and serum estradiol were measured in 11 premenopausal (Pre-M, 21 ± 1 yr), 13 perimenopausal (Peri-M, 49 ± 2 yr), and 10 postmenopausal (Post-M, 65 ± 7 yr) women. Measurements were made twice within the Pre-M group at their early follicular (EF, lower estradiol) and late follicular (LF, higher estradiol) phases. Shear-mediated dilation was induced by 3 min of hypercapnia (target PETCO2 + 10 mmHg from individual baseline) and was calculated as the percent rise in peak diameter relative to baseline diameter. ICA diameter and blood velocity were simultaneously measured by Doppler ultrasound. In Pre-M, shear-mediated dilation was higher during the LF phase than during the EF phase (P < 0.01). Comparing all groups, shear-mediated dilation was reduced across the menopausal transition (P < 0.01), and Pre-M during the LF phase showed the highest value (8.9 ± 1.4%) compared with other groups (Pre-M in EF, 6.4 ± 1.1%; Peri-M, 5.5 ± 1.3%; Post-M, 5.2 ± 1.9%, P < 0.05 for all). Shear-mediated dilation was positively correlated with serum estradiol even after adjustment of age (P < 0.01, r = 0.55, age-adjusted; P = 0.02, r = 0.35). Collectively, these data indicate that controlling the menstrual cycle phase is necessary for the cross-sectional assessments of shear-mediated dilation of the ICA in premenopausal women. Moreover, current findings suggest that a decline in cerebrovascular endothelial function may be partly related to the reduced circulating estrogen levels in peri- and postmenopausal women.NEW & NOTEWORTHY The present study evaluated the effects of the menstrual cycle and menopause stages on the shear-mediated dilation of the ICA, a potential index of cerebrovascular endothelial function, in pre-, peri-, and postmenopausal women. Shear-mediated dilation of the ICA was increased from the low- to high-estradiol phases in naturally cycling premenopausal women and was reduced with advancing menopause stages. Furthermore, lower estradiol was associated with reduced shear-mediated dilation of the ICA, independent of age.
Collapse
Affiliation(s)
- Erika Iwamoto
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Rintaro Sakamoto
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Wakako Tsuchida
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Kagawa, Japan
| | - Kotomi Yamazaki
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Tatsuki Kamoda
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Toru Neki
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Masaki Katayose
- School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Darren P Casey
- Department of Physical Therapy and Rehabilitation Science, Iowa City, Iowa.,Abboud Cardiovascular Research Center, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
43
|
Godo S, Shimokawa H. Gender Differences in Endothelial Function and Coronary Vasomotion Abnormalities. GENDER AND THE GENOME 2020. [DOI: 10.1177/2470289720957012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction: Structural and functional abnormalities of coronary microvasculature, referred to as coronary microvascular dysfunction (CMD), have been implicated in a wide range of cardiovascular diseases and have gained growing attention in patients with chest pain with no obstructive coronary artery disease, especially in females. The central mechanisms of coronary vasomotion abnormalities encompass enhanced coronary vasoconstrictive reactivity (ie, coronary spasm), reduced endothelium-dependent and -independent coronary vasodilator capacities, and increased coronary microvascular resistance. The 2 major endothelium-derived relaxing factors, nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) factors, modulate vascular tone in a distinct vessel size–dependent manner; NO mainly mediates vasodilatation of relatively large, conduit vessels, while EDH factors in small resistance vessels. Endothelium-dependent hyperpolarization–mediated vasodilatation is more prominent in female resistance arteries, where estrogens exert beneficial effects on endothelium-dependent vasodilatation via multiple mechanisms. In the clinical settings, therapeutic approaches targeting NO are disappointing for the treatment of various cardiovascular diseases, where endothelial dysfunction and CMD are substantially involved. Significance: In this review, we will discuss the current knowledge on the pathophysiology and molecular mechanisms of endothelial function and coronary vasomotion abnormalities from bench to bedside, with a special reference to gender differences. Results: Recent experimental and clinical studies have demonstrated distinct gender differences in endothelial function and coronary vasomotion abnormalities with major clinical implications. Moreover, recent landmark clinical trials regarding the management of stable coronary artery disease have questioned the benefit of percutaneous coronary intervention, supporting the importance of the coronary microvascular physiology. Conclusion: Further characterization and a better understanding of the gender differences in basic vascular biology as well as those in cardiovascular diseases are indispensable to improve health care and patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Shigeo Godo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
44
|
Qin P, Shi J, Cao L, Li X, Lou Y, Wang C, Ma J, Wang L, Peng X, Chen H, Xu S, Zhao P, Hu D, Hu F. Low-Density Lipoprotein Cholesterol/High-Density Lipoprotein Cholesterol Ratio and Carotid Intima-Media Thickness: A Cohort Study in China. Lipids 2020; 56:59-68. [PMID: 32856731 DOI: 10.1002/lipd.12274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 11/10/2022]
Abstract
Low-density lipoprotein cholesterol to high-density lipoprotein cholesterol (LDL-C/HDL-C) ratio may carry additional information and has been suggested as a better predictor for atherosclerosis progression and cardiovascular disease (CVD) than LDL-C and HDL-C alone. Therefore, we aim to explore the association between LDL-C/HDL-C ratio and high carotid intima-media thickness (CIMT) risk in a large Cohort in Beijing, China. This cohort study included 13,612 adults without high CIMT at first entry and who attended the baseline examination and at least one follow-up annual examination between 2009 and 2016. We used multivariable Cox regression to estimate hazard ratios (HR) with their 95% confidence intervals (CI) for the association between LDL-C/HDL-C ratio and risk of high CIMT. During 37,912 person-years of follow-up, 1996 (1268 men and 728 women) developed high CIMT. Compared with the first quartile of LDL-C/HDL-C ratio, the risk of high CIMT was significantly increased for the fourth quartile of LDL-C/HDL-C ratio (HR = 1.51, 95% CI: 1.29-1.78). We observed a significant association between LDL-C/HDL-C ratio and high CIMT risk comparing LDL-C/HDL-C ratio >2.78 with LDL-C/HDL-C ratio ≤2.78 and significant dose-response relationship between LDL-C/HDL-C ratio and risk of high CIMT. The restricted cubic spline showed a significant nonlinear association between LDL-C/HDL-C ratio and the risk of high CIMT (pnon-linearity = 0.009). We identify a significant association between LDL-C/HDL-C ratio and the risk of high CIMT in the Chinese Cohort study. Future studies are needed to evaluate the effectiveness of reducing LDL-C/HDL-C ratio treatment on the development of high CIMT.
Collapse
Affiliation(s)
- Pei Qin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, 518060, People's Republic of China
| | - Jing Shi
- Department of Epidemiology, College of Public Health, Harbin Medical University, No.157 Health Road, Harbin, Heilongjiang, 150000, People's Republic of China
| | - Liming Cao
- Department of Epidemiology, College of Public Health, Harbin Medical University, No.157 Health Road, Harbin, Heilongjiang, 150000, People's Republic of China
| | - Xue Li
- Department of Epidemiology, College of Public Health, Harbin Medical University, No.157 Health Road, Harbin, Heilongjiang, 150000, People's Republic of China
| | - Yanmei Lou
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing, 102200, People's Republic of China
| | - Changyi Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Jianping Ma
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Li Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Xiaolin Peng
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Hongen Chen
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Shan Xu
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, No.7 Huaming Road, Shenzhen, Guangdong, 102200, People's Republic of China
| | - Ping Zhao
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing, 102200, People's Republic of China
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, 518060, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, 518060, People's Republic of China
| |
Collapse
|
45
|
Masaki N, Ido Y, Yamada T, Yamashita Y, Toya T, Takase B, Hamburg NM, Adachi T. Endothelial Insulin Resistance of Freshly Isolated Arterial Endothelial Cells From Radial Sheaths in Patients With Suspected Coronary Artery Disease. J Am Heart Assoc 2020; 8:e010816. [PMID: 30885039 PMCID: PMC6475050 DOI: 10.1161/jaha.118.010816] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Endothelial insulin resistance is insulin‐insensitivity in the vascular endothelium and can be observed in experimental models. This study aimed to investigate endothelial insulin resistance in patients with suspected coronary artery disease. To this end, a novel method of obtaining freshly isolated arterial endothelial cells from a radial catheter sheath was developed. Methods and Results Freshly isolated arterial endothelial cells were retrieved from catheter sheaths placed in radial arteries for coronary angiography (n=69, patient age 64±12 years). The endothelial cells were divided into groups for incubation with or without insulin, vascular endothelial growth factor, or acetylcholine. The intensity of phosphorylated endothelial nitric oxide synthase at Ser1177 (p‐eNOS) was quantified by immunofluorescence microscopy. The percentage increase of insulin‐induced phosphorylated endothelial nitric oxide synthase correlated negatively with derivatives of reactive oxygen metabolites, an oxidative stress test (r=−0.348, n=53, P=0.011), E/E′, an index of left ventricular diastolic dysfunction in Doppler echocardiography (ρ=−0.374, n=49, P=0.008), and log‐transformed brain natriuretic peptide (r=−0.266, n=62, P=0.037). Furthermore, percentage increase of insulin‐induced p‐eNOS was an independent factor for the cardio‐ankle vascular index (standardized coefficient β=−0.293, n=42, P=0.021) in the multivariate regression analysis of adaptive least absolute shrinkage and selection operator. Conclusions Our results suggested that endothelial insulin resistance is associated with oxidative stress, left ventricular diastolic dysfunction, heart failure, and arterial stiffness.
Collapse
Affiliation(s)
- Nobuyuki Masaki
- 1 Department of Intensive Care Medicine National Defense Medical College Tokorozawa Japan
| | - Yasuo Ido
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Toshiyuki Yamada
- 3 Department of Cardiovascular Surgery Keio University Graduate School of Medicine Tokyo Japan
| | - Youhei Yamashita
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Takumi Toya
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| | - Bonpei Takase
- 1 Department of Intensive Care Medicine National Defense Medical College Tokorozawa Japan
| | - Naomi M Hamburg
- 4 The Whitaker Cardiovascular Institute Department of Medicine Boston University School of Medicine Boston MA
| | - Takeshi Adachi
- 2 Department of Cardiology National Defense Medical College Tokorozawa Japan
| |
Collapse
|
46
|
Stanojlović M, Guševac Stojanović I, Zarić M, Martinović J, Mitrović N, Grković I, Drakulić D. Progesterone Protects Prefrontal Cortex in Rat Model of Permanent Bilateral Common Carotid Occlusion via Progesterone Receptors and Akt/Erk/eNOS. Cell Mol Neurobiol 2020; 40:829-843. [PMID: 31865501 PMCID: PMC11448933 DOI: 10.1007/s10571-019-00777-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023]
Abstract
Sustained activation of pro-apoptotic signaling due to a sudden and prolonged disturbance of cerebral blood circulation governs the neurodegenerative processes in prefrontal cortex (PFC) of rats whose common carotid arteries are permanently occluded. The adequate neuroprotective therapy should minimize the activation of toxicity pathways and increase the activity of endogenous protective mechanisms. Several neuroprotectants have been proposed, including progesterone (P4). However, the underlying mechanism of its action in PFC following permanent bilateral occlusion of common carotid arteries is not completely investigated. We, thus herein, tested the impact of post-ischemic P4 treatment (1.7 mg/kg for seven consecutive days) on previously reported aberrant neuronal morphology and amount of DNA fragmentation, as well as the expression of progesterone receptors along with the key elements of Akt/Erk/eNOS signal transduction pathway (Bax, Bcl-2, cytochrome C, caspase 3, PARP, and the level of nitric oxide). The obtained results indicate that potential amelioration of histological changes in PFC might be associated with the absence of activation of Bax/caspase 3 signaling cascade and the decline of DNA fragmentation. The study also provides the evidence that P4 treatment in repeated regiment of administration might be effective in neuronal protection against ischemic insult due to re-establishment of the compromised action of Akt/Erk/eNOS-mediated signaling pathway and the upregulation of progesterone receptors.
Collapse
Affiliation(s)
- Miloš Stanojlović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Ivana Guševac Stojanović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Marina Zarić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Jelena Martinović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia
| | - Dunja Drakulić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, Belgrade, 11001, Republic of Serbia.
| |
Collapse
|
47
|
Endothelial dysfunction induced by hydroxyl radicals – the hidden face of biodegradable Fe-based materials for coronary stents. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110938. [DOI: 10.1016/j.msec.2020.110938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 02/14/2020] [Accepted: 04/05/2020] [Indexed: 12/11/2022]
|
48
|
Lou Y, Li X, Cao L, Qin P, Shi J, Zhang Y, Wang C, Ma J, Wang L, Peng X, Chen H, Xu S, Hu F, Zhao Y, Zhao P. LDL-cholesterol to HDL-cholesterol ratio discordance with lipid parameters and carotid intima-media thickness: a cohort study in China. Lipids Health Dis 2020; 19:141. [PMID: 32552893 PMCID: PMC7302368 DOI: 10.1186/s12944-020-01324-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
Background The discordance of the low-density lipoprotein cholesterol-to-high-density lipoprotein cholesterol (LDL-C/HDL-C) ratio with alterative lipid parameters may explain the inconsistent association of CIMT with the LDL-C/HDL-C ratio. Therefore, this study aimed to explore the associations between LDL-C/HDL-C ratio discordance with alternative lipid parameters and elevated carotid intima-media thickness (CIMT) risk in a large cohort in Beijing, China. Methods In total, 13,612 adults who didn’t have elevated CIMT at baseline and who participated in at least one follow-up of annual examination between 2009 and 2016 were included in this cohort study. A multivariable Cox regression model was utilized to evaluate the associations of discordance of the LDL-C/HDL-C ratio with TC, TGs, LDL-C and HDL-C with elevated CIMT risk. Results During 37,999 person-years of follow-up, 2004 individuals (1274 men and 730 women) developed elevated CIMT. Among individuals with normal TC and TGs, 16.6 and 15.2% individuals had a discordantly high LDL-C/HDL-C ratio, respectively, and the risk of elevated CIMT increased by 1.54 (95% CI 1.33, 1.77) and 1.53 (95% CI 1.33, 1.76), respectively, comparing to individuals with a concordantly low LDL-C/HDL-C ratio. A high LDL-C/HDL-C ratio could significantly increase elevated CIMT risk regardless of discordance/concordance with LDL-C and HDL-C (P < 0.001). A low LDL-C/HDL-C ratio with discordantly normal HDL-C and high LDL-C (13.2% of individuals) had a 32% (HR = 1.32, 95% CI 1.11, 1.57) higher risk of elevated CIMT than concordantly low LDL-C and normal HDL-C. Sensitivity analysis by excluding CIMT developed in the first 2 years follow-up further confirmed the above results. Conclusions A high LDL-C/HDL-C ratio could significantly increase elevated CIMT risk regardless of discordance/concordance with TC, TGs, LDL-C and HDL-C Even a low LDL-C/HDL-C ratio with discordantly high LDL-C and normal HDL-C could also significantly increase CIMT risk. Individuals should maintain both the LDL-C/HDL-C ratio and LDL-C at normal levels to prevent elevated CIMT.
Collapse
Affiliation(s)
- Yanmei Lou
- Department of Health Management, Beijing Xiao Tang Shan Hospital, Beijing, People's Republic of China
| | - Xue Li
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Liming Cao
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Pei Qin
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Jing Shi
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Yanyan Zhang
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Changyi Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Jianping Ma
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Li Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Xiaolin Peng
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Hongen Chen
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Shan Xu
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic disease, Shenzhen, Guangdong, People's Republic of China
| | - Fulan Hu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China.
| | - Yashuang Zhao
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China.
| | - Ping Zhao
- Department of Health Management, Beijing Xiao Tang Shan Hospital, Beijing, People's Republic of China.
| |
Collapse
|
49
|
Harvey RE, Ranadive SM, Limberg JK, Baker SE, Nicholson WT, Curry TB, Barnes JN, Joyner MJ. Forearm vasodilatation to a β 2 -adrenergic receptor agonist in premenopausal and postmenopausal women. Exp Physiol 2020; 105:886-892. [PMID: 32170888 DOI: 10.1113/ep088452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/10/2020] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the central question of this study? What is the role of β2 -adrenergic receptor (β2 AR) vasodilatation in older postmenopausal women as compared to premenopausal women and the role of nitric oxide (NO) in β2 AR-mediated vasodilatation in both groups of women? What is the main finding and its importance? β2 AR responsiveness is blunted in postmenopausal women compared to young premenopausal women. Additionally, NO may contribute to β2 AR-mediated vasodilatation in young premenopausal women. ABSTRACT β2 -Adrenergic receptor (β2 AR)-mediated vasodilatation, which is partially dependent on nitric oxide (NO) formation, is blunted in men at risk for developing hypertension. However, the role of β2 AR vasodilatation in hypertension pathophysiology in ageing postmenopausal women is unclear. Therefore, the goals of this study were to determine if forearm vasodilatation to the selective β2 AR agonist terbutaline is blunted in older postmenopausal women (59 ± 4 years) compared to young premenopausal women (27 ± 3 years) and to assess NO contribution to β2 AR-mediated vasodilatation in both groups of women. Forearm blood flow (FBF) and forearm vascular conductance (FVC) were measured using venous occlusion plethysmography at baseline and during intra-arterial infusions of terbutaline at 0.1-2.0 µg (100 ml tissue)-1 min-1 with and without the NO synthase inhibitor l-NG -monomethylarginine (l-NMMA). Mean arterial pressure was significantly greater in postmenopausal women than in young women at baseline (P = 0.01). Baseline FBF and FVC did not differ between young and postmenopausal women (P > 0.05) and rose significantly within each group during terbutaline infusion (P < 0.05). There were significant group × dose interactions for FBF (P = 0.01) and FVC (P = 0.001), indicating vasodilator responses were lower in postmenopausal women. In young women, FVC response to the highest dose of terbutaline tended to be lower with l-NMMA co-infusion vs. without l-NMMA (P = 0.05). There were no significant decreases in FBF or FVC responses to terbutaline in postmenopausal women with l-NMMA co-infusion (P > 0.05 for all). These data suggest that β2 AR responsiveness is blunted in postmenopausal women compared to young premenopausal women, and that NO may contribute to β2 AR-mediated vasodilatation in young premenopausal women.
Collapse
Affiliation(s)
- Ronée E Harvey
- Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sushant M Ranadive
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA.,Department of Kinesiology, University of Maryland, College Park, MD, USA
| | - Jacqueline K Limberg
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA.,Department of Nutrition and Exercise Physiology, University of Missouri Columbia, MO, USA
| | - Sarah E Baker
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | | | - Timothy B Curry
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Jill N Barnes
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA.,Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
50
|
Triggle CR, Ding H, Marei I, Anderson TJ, Hollenberg MD. Why the endothelium? The endothelium as a target to reduce diabetes-associated vascular disease. Can J Physiol Pharmacol 2020; 98:415-430. [PMID: 32150686 DOI: 10.1139/cjpp-2019-0677] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the past 66 years, our knowledge of the role of the endothelium in the regulation of cardiovascular function and dysfunction has advanced from the assumption that it is a single layer of cells that serves as a barrier between the blood stream and vascular smooth muscle to an understanding of its role as an essential endocrine-like organ. In terms of historical contributions, we pay particular credit to (1) the Canadian scientist Dr. Rudolf Altschul who, based on pathological changes in the appearance of the endothelium, advanced the argument in 1954 that "one is only as old as one's endothelium" and (2) the American scientist Dr. Robert Furchgott, a 1998 Nobel Prize winner in Physiology or Medicine, who identified the importance of the endothelium in the regulation of blood flow. This review provides a brief history of how our knowledge of endothelial function has advanced and now recognize that the endothelium produces a plethora of signaling molecules possessing paracrine, autocrine, and, arguably, systemic hormone functions. In addition, the endothelium is a therapeutic target for the anti-diabetic drugs metformin, glucagon-like peptide I (GLP-1) receptor agonists, and inhibitors of the sodium-glucose cotransporter 2 (SGLT2) that offset the vascular disease associated with diabetes.
Collapse
Affiliation(s)
- Chris R Triggle
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Hong Ding
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Isra Marei
- Departments of Pharmacology and Medical Education, Weill Cornell Medical College, Doha, Qatar
| | - Todd J Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| | - Morley D Hollenberg
- Inflammation Research Network, Snyder Institute for Chronic Disease, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada.,Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada.,Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|