1
|
Brong A, Kontrogianni-Konstantopoulos A. Sex Chromosomes and Sex Hormones: Dissecting the Forces That Differentiate Female and Male Hearts. Circulation 2025; 151:474-489. [PMID: 39960989 PMCID: PMC11839176 DOI: 10.1161/circulationaha.124.069493] [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] [Indexed: 02/21/2025]
Abstract
The heart is a highly sex-biased organ, as sex shapes innumerable aspects of heart health and disease. Sex chromosomes and sex hormones -testosterone, progesterone, and estrogen- establish and perpetuate the division between male and female myocardium. Of these differentiating factors, the insulating effects of estrogen have been rigorously interrogated and reviewed, whereas the influence of sex chromosomes, testosterone, and progesterone remains in dispute or ill-defined. Here, we synthesize growing evidence that sex chromosomes and sex hormones substantially bias heart form, function, and dysfunction in a context-dependent fashion. The discrete protective functions ascribed to each of the 3 estrogen receptors are also enumerated. Subsequently, we overview obstacles that have historically discouraged the inclusion of female subjects in basic science such as the impact of the female estrus cycle and reproductive senescence on data reliability and reproducibility. Furthermore, we weigh the utility of several common strategies to intercept and rescue sex-specific protection. Last, we warn of common compounds in animal chow and cell culture that interfere with estrogen signaling. In sum, we survey the controversies and challenges that stem from sex-inclusive cardiovascular research, comparing the possible causes of cardiac sex bias, elucidating sex chromosome or hormone-dependent processes in the heart, describing common lapses that imperil female and male cell and animal work, and illuminating facets of the female heart yet unexplored or still uncertain.
Collapse
Affiliation(s)
- Annie Brong
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Aikaterini Kontrogianni-Konstantopoulos
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Shams AA, Vesal S, Karoii DH, Vesali S, Alizadeh A, Shahhoseini M. Paternal trans fatty acid and vitamin E diet affect the expression pattern of androgen signaling pathway genes in the testis of rat offspring. Theriogenology 2025; 231:1-10. [PMID: 39378727 DOI: 10.1016/j.theriogenology.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Numerous studies have shown that an improper diet in parents has a negative impact on offspring's health. Furthermore, the negative effects of trans fatty acids (TFA) in maternal diets on fertility and health and their impact on future generations have been documented. However, there is limited research on the negative effects of TFA in paternal diets on male children. The current work used qRT-PCR to investigate the effects of trans fatty acids and vitamin E in the paternal diet on the expression pattern of androgen signaling pathway genes such as STAR, CYP11a1, HSD3B, SRD5a2, and SCARB1 in offspring testes. In this experiment, parental rats were randomly separated into four groups, each with ten father rats, and were fed for eight weeks (60 days) as follows. 1: Standard diet group plus liquid sunflower oil (control). 2: Standard diet group containing trans fatty acids (CTH). 3: The regular diet group received 2.5 times the recommended quantity of vitamin E supplement. 4: Standard diet group with vitamin E and trans fatty acid supplementation (ETH). The testis tissue samples from 35 offspring were then used. Following RNA extraction from tissues and cDNA synthesis, quantitative real-time PCR was used to evaluate the expression levels of androgen signaling pathway genes such as STAR, CYP11A1, HSD3B, SCARB1, and SRD5A2. Our findings showed that the expression of CYP11A1 was considerably reduced in the progeny of paternal rats given ETH compared to the CTH group. The expression levels of the STAR gene were significantly lower in the progeny of paternal rats administered TFA, ETH, and vitamin E compared to the controls. Although the CTH group had lower SCARB1 expression than the other groups, the difference was not statistically significant. Paternal vitamin E consumption substantially affected SRD5A2 expression when compared to offspring of paternal rats fed vitamin E + trans fatty acid or those fed a conventional diet containing trans fatty acid. Furthermore, the vitamin E group showed a statistically significant increase in HSD3B expression compared to the other groups. Bioinformatics analyses, such as protein-protein interaction networks and gene ontology term enrichment, revealed that these genes play roles in lipid biosynthesis, hormone metabolism, male sex differentiation, reproductive development, and steroid biosynthesis. Our data indicate that paternal trans fatty acid consumption influences the expression of particular androgen signaling pathway genes in offspring testis, with vitamin E potentially mitigating some of these effects.
Collapse
Affiliation(s)
- Amir Abbas Shams
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Soheil Vesal
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science University of Tehran, Tehran, Iran
| | - Samira Vesali
- Department of Basic and Population Based Studies in NCD, Reproductive Epidemiology Research Centre, Royan Institute, ACECR, Tehran, Iran
| | - AliReza Alizadeh
- Department of Embryology, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Shahhoseini
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran; Department of Cell and Molecular Biology, School of Biology, College of Science University of Tehran, Tehran, Iran; Department of Genetics, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Lian X, Bai Y, Du P, Jing Z, Gao J, Liu F, Hu J, Xi Y. Causal influences of testosterone on brain structure change rate: A sex-stratified Mendelian randomization study. J Steroid Biochem Mol Biol 2025; 245:106629. [PMID: 39481491 DOI: 10.1016/j.jsbmb.2024.106629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
The impact of testosterone levels on changes in brain structure has been reported. However, it is still unclear which specific brain region could be affected. This study approached Mendelian randomization method to reveal the causal relationship between testosterone levels and the rate of longitudinal structural changes in the brain. The testosterone-related GWAS data were determined from 425,097 European participants. The GWAS data on the rate of longitudinal structural changes in the brain came from the ENIGMA consortium, which included 15,640 all-age participants from 40 longitudinal cohorts. The inverse variance weighted was considered as the main estimate, MR Egger and weighted median methods were used to supplement IVW. A positive correlation was found between total testosterone levels and bioavailable testosterone levels in women and age-independent longitudinal changes in cerebral WM and surface area. The sex hormone-binding globulin levels were found a negative correlation with age-dependent longitudinal structural changes of cortical GM in men. Additionally, we also found that the bioavailable testosterone level in males was negatively associated with the quadratic age-dependent longitudinal change rate in the globus pallidum. We also found estradiol levels and sex hormone-binding globulin levels were negatively associated with the quadratic age-dependent longitudinal change rate of total brain in men. Moreover, we found a positive correlation between total testosterone levels and linear age-dependent longitudinal changes in the hippocampus in both males and females. The testosterone levels in different genders may have varying degrees of causal effects on the structural changes of brain regions. These findings provide evidence for the influence of the brain glandular axis on brain structure, particularly during female brain development.
Collapse
Affiliation(s)
- Xin Lian
- Department of Obstetrics and Gynecology, People's Hospital of Linyi County, Yuncheng Central Hospital of Shanxi Medical University, 1125 Fuxi Street, Yuncheng 044100, China
| | - Yaqi Bai
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Pengyang Du
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Zhinan Jing
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Jimi Gao
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Fan Liu
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Jingjing Hu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China.
| | - Yujia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, Male Reproductive Medicine Center, 382 Wuyi Road, Taiyuan 030001, China.
| |
Collapse
|
4
|
Liu CY, Tsai TY, Liu TH, Chang TC, Chen YW, Tsao CW. Lactiplantibacillus plantarum 1008 Promotes Reproductive Function and Cognitive Activity in Aged Male Mice with High-Fat-Diet-Induced Obesity by Altering Metabolic Parameters and Alleviating Testicular Oxidative Damage, Inflammation and Apoptosis. Antioxidants (Basel) 2024; 13:1498. [PMID: 39765826 PMCID: PMC11673844 DOI: 10.3390/antiox13121498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The effects of Lactiplantibacillus plantarum 1008 (LP1008) on age-related cognitive impairment and skeletal muscle atrophy have been reported previously. However, its role in obesity- and age-related hypogonadism has yet to be explored. This study investigates the therapeutic efficacy of low- and high-dose LP1008 in a high-fat-diet-fed male mouse model. Mice at 37 weeks of age were fed a standard diet (n = 8) or a 45% high-fat diet for 28 weeks, and the high-fat-diet-fed mice were divided into vehicle, low-dose and high-dose LP1008 groups (n = 8 per group) on the basis of the treatment administered for an additional 8 weeks. We found that LP1008 suppressed the increases in total cholesterol levels and liver function parameters and alleviated histological changes in the brain, ileum, gastrocnemius muscle and testes. In terms of reproductive function, LP1008 attenuated the decreases in sperm quality, sperm maturity, testosterone levels and levels of enzymes involved in testosterone biosynthesis. Furthermore, LP1008 altered impairments in spatial learning and memory and induced slight alterations in the gut microbiota. Moreover, LP1008 exerted antioxidant, anti-inflammatory and anti-apoptotic effects in aged, obese male mice. LP1008 reversed diet-induced obesity, age-related reproductive dysfunction and pathological damage by increasing testosterone levels and altering the gut microbiome through the regulation of mediators involved in oxidative stress, apoptosis and inflammation.
Collapse
Affiliation(s)
- Chin-Yu Liu
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (C.-Y.L.); (T.-C.C.); (Y.-W.C.)
| | - Tsung-Yu Tsai
- Department of Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.T.); (T.-H.L.)
| | - Te-Hua Liu
- Department of Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.T.); (T.-H.L.)
| | - Ting-Chia Chang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (C.-Y.L.); (T.-C.C.); (Y.-W.C.)
| | - Yi-Wen Chen
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (C.-Y.L.); (T.-C.C.); (Y.-W.C.)
| | - Chih-Wei Tsao
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
5
|
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
|
6
|
Wang Z, Zhang G, Hu S, Fu M, Zhang P, Zhang K, Hao L, Chen S. Research progress on the protective effect of hormones and hormone drugs in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 176:116764. [PMID: 38805965 DOI: 10.1016/j.biopha.2024.116764] [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: 02/21/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Ischemic heart disease (IHD) is a condition where the heart muscle does not receive enough blood flow, leading to cardiac dysfunction. Restoring blood flow to the coronary artery is an effective clinical therapy for myocardial ischemia. This strategy helps lower the size of the myocardial infarction and improves the prognosis of patients. Nevertheless, if the disrupted blood flow to the heart muscle is restored within a specific timeframe, it leads to more severe harm to the previously deprived heart tissue. This condition is referred to as myocardial ischemia/reperfusion injury (MIRI). Until now, there is a dearth of efficacious strategies to prevent and manage MIRI. Hormones are specialized substances that are produced directly into the circulation by endocrine organs or tissues in humans and animals, and they have particular effects on the body. Hormonal medications utilize human or animal hormones as their active components, encompassing sex hormones, adrenaline medications, thyroid hormone medications, and others. While several studies have examined the preventive properties of different endocrine hormones, such as estrogen and hormone analogs, on myocardial injury caused by ischemia-reperfusion, there are other hormone analogs whose mechanisms of action remain unexplained and whose safety cannot be assured. The current study is on hormones and hormone medications, elucidating the mechanism of hormone pharmaceuticals and emphasizing the cardioprotective effects of different endocrine hormones. It aims to provide guidance for the therapeutic use of drugs and offer direction for the examination of MIRI in clinical therapy.
Collapse
Affiliation(s)
- Zhongyi Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Gaojiang Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Meilin Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Pingyuan Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kuo Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Sichong Chen
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
7
|
Topriceanu CC, Shah M, Webber M, Chan F, Shiwani H, Richards M, Schott J, Chaturvedi N, Moon JC, Hughes AD, Hingorani AD, O'Regan DP, Captur G. APOE ε4 carriage associates with improved myocardial performance from adolescence to older age. BMC Cardiovasc Disord 2024; 24:172. [PMID: 38509472 PMCID: PMC10956279 DOI: 10.1186/s12872-024-03808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Although APOE ε4 allele carriage confers a risk for coronary artery disease, its persistence in humans might be explained by certain survival advantages (antagonistic pleiotropy). METHODS Combining data from ~ 37,000 persons from three older age British cohorts (1946 National Survey of Health and Development [NSHD], Southall and Brent Revised [SABRE], and UK Biobank) and one younger age cohort (Avon Longitudinal Study of Parents and Children [ALSPAC]), we explored whether APOE ε4 carriage associates with beneficial or unfavorable left ventricular (LV) structural and functional metrics by echocardiography and cardiovascular magnetic resonance (CMR). RESULTS Compared to the non-APOE ε4 group, APOE ε4 carriers had similar cardiac phenotypes in terms of LV ejection fraction, E/e', posterior wall and interventricular septal thickness, and LV mass. However, they had improved myocardial performance resulting in greater LV stroke volume generation per 1 mL of myocardium (higher myocardial contraction fraction). In NSHD (n = 1467) and SABRE (n = 1187), ε4 carriers had a 4% higher MCF (95% CI 1-7%, p = 0.016) using echocardiography. Using CMR data, in UK Biobank (n = 32,972), ε4 carriers had a 1% higher MCF 95% (CI 0-1%, p = 0.020) with a dose-response relationship based on the number of ε4 alleles. In addition, UK Biobank ε4 carriers also had more favorable radial and longitudinal strain rates compared to non APOE ε4 carriers. In ALSPAC (n = 1397), APOE ε4 carriers aged < 24 years had a 2% higher MCF (95% CI 0-5%, p = 0.059). CONCLUSIONS By triangulating results in four independent cohorts, across imaging modalities (echocardiography and CMR), and in ~ 37,000 individuals, our results point towards an association between ε4 carriage and improved cardiac performance in terms of LV MCF. This potentially favorable cardiac phenotype adds to the growing number of reported survival advantages attributed to the pleiotropic effects APOE ε4 carriage that might collectively explain its persistence in human populations.
Collapse
Affiliation(s)
- Constantin-Cristian Topriceanu
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
- Cardiology Department, Centre for Inherited Heart Muscle Conditions, The Royal Free Hospital, Pond Street, Hampstead, London, UK
| | - Mit Shah
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Science, Imperial College London, London, UK
| | - Matthew Webber
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Fiona Chan
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Hunain Shiwani
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
| | - Marcus Richards
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Jonathan Schott
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Nishi Chaturvedi
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - James C Moon
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
| | - Alun D Hughes
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Aroon D Hingorani
- UCL Institute of Cardiovascular Science, University College London, London, UK
- BHF Research Accelerator, University College London, London, UK
- Health Data Research, University College London, London, UK
| | - Declan P O'Regan
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Science, Imperial College London, London, UK
| | - Gabriella Captur
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK.
- UCL Institute of Cardiovascular Science, University College London, London, UK.
- Cardiac MRI Unit, Barts Heart Centre, London, UK.
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK.
- Cardiology Department, Centre for Inherited Heart Muscle Conditions, The Royal Free Hospital, Pond Street, Hampstead, London, UK.
| |
Collapse
|
8
|
Matthews EL, Guers JJ, Ramick MG, Hosick PA. Inverse Association between Exercising Blood Pressure Response and Left Ventricular Chamber Size and Mass in Women Who Habitually Resistance Train. Healthcare (Basel) 2024; 12:353. [PMID: 38338238 PMCID: PMC10855375 DOI: 10.3390/healthcare12030353] [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: 12/04/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Exercise is a major modifiable lifestyle factor that leads to temporarily increased systolic blood pressure (SBP), which is thought to influence left ventricular mass normalized to body surface area (LVM/BSA). This relationship has never been studied in women who habitually perform resistance exercise. PURPOSE To determine if a direct correlation exists between the SBP response to resistance exercise (change from rest; eSBP) and LVM/BSA in young healthy women who habitually resistance train. METHODS Leg extension resistance exercise was performed while continuously monitoring blood pressure using finger plethysmography. LVM was estimated using echocardiography. Data are shown as mean ± SD. RESULTS Thirty-one women participated (age 23 ± 3 years, height 164 ± 7 cm, body mass 63.7 ± 10.3 kg). Resting SBP (110 ± 8 mmHg, r = 0.355, p = 0.049) was shown to be directly correlated to LVM/BSA (72.0 ± 28.4 g/m2). Conversely, eSBP (30.8 ± 14.6 ∆mmHg, r = -0.437, p = 0.014) was inversely related to LVM/BSA. eSBP was not correlated to interventricular septum width (0.88 ± 0.12 cm, r = -0.137, p = 0.463) or posterior wall thickness (0.91 ± 0.15 cm, r = -0.084, p = 0.654). eSBP was inversely related to left ventricle internal diameter during diastole (LVIDd) (4.25 ± 0.33 cm, r = -0.411, p = 0.021). CONCLUSION Counter to the hypothesis, these data suggest an inverse association between eSBP during resistance exercise and LVM/BSA in healthy young women who resistance train. This relationship is due to a smaller LVIDd with greater eSBP.
Collapse
Affiliation(s)
- Evan L. Matthews
- Department of Exercise Science and Physical Education, Montclair State University, Montclair, NJ 07043, USA;
| | - John J. Guers
- Department of Biology, Behavioral Neuroscience and Health Sciences, Rider University, Lawrenceville, NJ 08648, USA;
| | - Meghan G. Ramick
- Department of Kinesiology, West Chester University, West Chester, PA 19383, USA;
| | - Peter A. Hosick
- Department of Exercise Science and Physical Education, Montclair State University, Montclair, NJ 07043, USA;
| |
Collapse
|
9
|
Zhao J, He C, Fan X, Wang L, Zhao L, Liu H, Shen W, Jiang S, Pei K, Gao J, Qi Y, Liu Y, Zhao J, Zhang R, Lu C, Tong J, Huai J. Tripeptidyl peptidase II coordinates the homeostasis of calcium and lipids in the central nervous system and its depletion causes presenile dementia in female mice through calcium/lipid dyshomeostasis-induced autophagic degradation of CYP19A1. Theranostics 2024; 14:1390-1429. [PMID: 38389851 PMCID: PMC10879859 DOI: 10.7150/thno.92571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Tripeptidyl peptidase II (TPP2) has been proven to be related to human immune and neurological diseases. It is generally considered as a cytosolic protein which forms the largest known protease complex in eukaryotic cells to operate mostly downstream of proteasomes for degradation of longer peptides. However, this canonical function of TPP2 cannot explain its role in a wide variety of biological and pathogenic processes. The mechanistic interrelationships and hierarchical order of these processes have yet to be clarified. Methods: Animals, cells, plasmids, and viruses established and/or used in this study include: TPP2 knockout mouse line, TPP2 conditional knockout mouse lines (different neural cell type oriented), TRE-TPP2 knockin mouse line on the C57BL/6 background; 293T cells with depletion of TPP2, ATF6, IRE1, PERK, SYVN1, UCHL1, ATG5, CEPT1, or CCTα, respectively; 293T cells stably expressing TPP2, TPP2 S449A, TPP2 S449T, or CCTα-KDEL proteins on the TPP2-depleted background; Plasmids for eukaryotic transient expression of rat CYP19A1-Flag, CYP19A1 S118A-Flag, CYP19A1 S118D-Flag, Sac I ML GFP Strand 11 Long, OMMGFP 1-10, G-CEPIA1er, GCAMP2, CEPIA3mt, ACC-GFP, or SERCA1-GFP; AAV2 carrying the expression cassette of mouse CYP19A1-3 X Flag-T2A-ZsGreen. Techniques used in this study include: Flow cytometry, Immunofluorescence (IF) staining, Immunohistochemical (IHC) staining, Luxol fast blue (LFB) staining, β-galactosidase staining, Lipid droplet (LD) staining, Calcium (Ca2+) staining, Stimulated emission depletion (STED) imaging, Transmission electron microscopic imaging, Two-photon imaging, Terminal deoxynucleotidyl transferase (TdT) dUTP nick-end Labeling (TUNEL) assay, Bromodeoxyuridine (BrdU) assay, Enzymatic activity assay, Proximity ligation assay (PLA), In vivo electrophysiological recording, Long-term potentiation (LTP) recording, Split-GFP-based mitochondria-associated membrane (MAM) detection, Immunoprecipitation (IP), Cellular fractionation, In situ hybridization, Semi-quantitative RT-PCR, Immunoblot, Mass spectrometry-based lipidomics, metabolomics, proteomics, Primary hippocampal neuron culture and Morris water maze (MWM) test. Results: We found that TPP2, independent of its enzymatic activity, plays a crucial role in maintaining the homeostasis of intracellular Ca2+ and phosphatidylcholine (PC) in the central nervous system (CNS) of mice. In consistence with the critical importance of Ca2+ and PC in the CNS, TPP2 gene ablation causes presenile dementia in female mice, which is closely associated with Ca2+/PC dysregulation-induced endoplasmic reticulum (ER) stress, abnormal autophagic degradation of CYP19A1 (aromatase), and estrogen depletion. This work therefore uncovers a new role of TPP2 in lipogenesis and neurosteroidogenesis which is tightly related to cognitive function of adult female mice. Conclusion: Our study reveals a crucial role of TPP2 in controlling homeostasis of Ca2+ and lipids in CNS, and its deficiency causes sexual dimorphism in dementia. Thus, this study is not only of great significance for elucidating the pathogenesis of dementia and its futural treatment, but also for interpreting the role of TPP2 in other systems and their related disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Chengtong He
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Liao Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Hui Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Wujun Shen
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Sanwei Jiang
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
| | - Kaixuan Pei
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jingjing Gao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yawei Qi
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yang Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Junqiang Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Ruiling Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Chengbiao Lu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
- Senior author for electrophysiological experiments and related analysis
| | - Jia Tong
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| |
Collapse
|
10
|
Hunter SK, S Angadi S, Bhargava A, Harper J, Hirschberg AL, D Levine B, L Moreau K, J Nokoff N, Stachenfeld NS, Bermon S. The Biological Basis of Sex Differences in Athletic Performance: Consensus Statement for the American College of Sports Medicine. Med Sci Sports Exerc 2023; 55:2328-2360. [PMID: 37772882 DOI: 10.1249/mss.0000000000003300] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
ABSTRACT Biological sex is a primary determinant of athletic performance because of fundamental sex differences in anatomy and physiology dictated by sex chromosomes and sex hormones. Adult men are typically stronger, more powerful, and faster than women of similar age and training status. Thus, for athletic events and sports relying on endurance, muscle strength, speed, and power, males typically outperform females by 10%-30% depending on the requirements of the event. These sex differences in performance emerge with the onset of puberty and coincide with the increase in endogenous sex steroid hormones, in particular testosterone in males, which increases 30-fold by adulthood, but remains low in females. The primary goal of this consensus statement is to provide the latest scientific knowledge and mechanisms for the sex differences in athletic performance. This review highlights the differences in anatomy and physiology between males and females that are primary determinants of the sex differences in athletic performance and in response to exercise training, and the role of sex steroid hormones (particularly testosterone and estradiol). We also identify historical and nonphysiological factors that influence the sex differences in performance. Finally, we identify gaps in the knowledge of sex differences in athletic performance and the underlying mechanisms, providing substantial opportunities for high-impact studies. A major step toward closing the knowledge gap is to include more and equitable numbers of women to that of men in mechanistic studies that determine any of the sex differences in response to an acute bout of exercise, exercise training, and athletic performance.
Collapse
Affiliation(s)
- Sandra K Hunter
- Exercise Science Program, Department of Physical Therapy, and Athletic and Human Performance Center, Marquette University, Milwaukee, WI
| | | | - Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for Reproductive Sciences, University of California, San Francisco, CA
| | - Joanna Harper
- Loughborough University, Loughborough, UNITED KINGDOM
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, and Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SWEDEN
| | - Benjamin D Levine
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, and the Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kerrie L Moreau
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, and Eastern Colorado Health Care System, Geriatric Research Education and Clinical Center, Aurora, CO
| | - Natalie J Nokoff
- Department of Pediatrics, Section of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Nina S Stachenfeld
- The John B. Pierce Laboratory and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Stéphane Bermon
- Health and Science Department, World Athletics, Monaco and the LAMHESS, University Côte d'Azur, Nice, FRANCE
| |
Collapse
|
11
|
Guo F, Mao S, Long Y, Zhou B, Gao L, Huang H. The Influences of Perinatal Androgenic Exposure on Cardiovascular and Metabolic Disease of Offspring of PCOS. Reprod Sci 2023; 30:3179-3189. [PMID: 37380913 DOI: 10.1007/s43032-023-01286-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: 01/11/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.
Collapse
Affiliation(s)
- Fei Guo
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Suqing Mao
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yuhang Long
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Bokang Zhou
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ling Gao
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hefeng Huang
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
12
|
Chen D, Lee C, Tsai M, Hsieh M, Chuang C, Pang S, Chen S, Tseng C, Chang S, Chu P, Hsieh I, Wu VC, Huang W. Cancer Therapy-Related Cardiac Dysfunction in Patients With Prostate Cancer Undergoing Androgen Deprivation Therapy. J Am Heart Assoc 2023; 12:e030447. [PMID: 37750600 PMCID: PMC10727237 DOI: 10.1161/jaha.123.030447] [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: 04/03/2023] [Accepted: 08/24/2023] [Indexed: 09/27/2023]
Abstract
Background The risk of cardiac dysfunction for patients with prostate cancer undergoing androgen deprivation therapy (ADT) in the real-world setting remains unclear. Methods and Results A total of 1120 patients with prostate cancer and a baseline echocardiography scan were identified from Chang Gung Research Database between January 1, 2001 and December 31, 2019. Patients were treated with gonadotropin-releasing hormone agonist therapy, gonadotropin-releasing hormone antagonist therapy, or bilateral orchiectomy. Changes in left ventricular ejection fraction (LVEF) were further assessed in 421 patients using repeated measurements of LVEF before and during ADT treatment. The incidence of cancer therapy-related cardiac dysfunction (CT-RCD) was evaluated and defined as a ≥10% absolute decline in LVEF from baseline to a value of <53%. Among 421 patients undergoing ADT, LVEF declined from 66.3±11.3% to 62.5±13.6% (95% CI of mean difference: -5.0% to -2.7%) after a mean follow-up period of 1.6±0.8 years. CT-RCD occurred in 58 patients (13.7%) with a nadir LVEF of 40.3±9.1% after ADT. Lower baseline LVEF was significantly associated with CT-RCD (odds ratio, 1.07 [95% CI, 1.04-1.10]). The area under the curve of baseline LVEF for discriminating CT-RCD was 75.6%, with the corresponding optimal cutoff value of 64.5% (sensitivity, 79.3%; specificity, 67.2%). Conclusions ADT with gonadotropin-releasing hormone agonist therapy, gonadotropin-releasing hormone antagonist therapy, and bilateral orchiectomy were associated with an increased risk of CT-RCD in patients with prostate cancer. In addition, lower baseline LVEF was a significant predictor of CT-RCD in patients with prostate cancer undergoing treatment with ADT.
Collapse
Affiliation(s)
- Dong‐Yi Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Cheng‐Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Ming‐Lung Tsai
- Division of CardiologyNew Taipei Municipal TuCheng HospitalNew Taipei CityTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Ming‐Jer Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Cheng‐Keng Chuang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
| | - See‐Tong Pang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
| | - Shao‐Wei Chen
- Chang Gung University College of MedicineTaoyuanTaiwan
- Department of Thoracic and Cardiovascular SurgeryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
- Center for Big Data Analytics and Statistics, Department of Medical Research and Development, Chang Gung Memorial Hospital at LinkouTaoyuanTaiwan
| | - Chi‐Nan Tseng
- Chang Gung University College of MedicineTaoyuanTaiwan
- Department of Thoracic and Cardiovascular SurgeryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
| | - Shang‐Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
| | - Pao‐Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - I‐Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Victor Chien‐Chia Wu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
- Chang Gung University College of MedicineTaoyuanTaiwan
| | - Wen‐Kuan Huang
- Chang Gung University College of MedicineTaoyuanTaiwan
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at LinkouChang Gung University College of MedicineTaoyuanTaiwan
| |
Collapse
|
13
|
do Val Lima PR, Ronconi KS, Morra EA, Rodrigues PL, Ávila RA, Merlo E, Graceli JB, Simões MR, Stefanon I, Ribeiro Júnior RF. Testosterone deficiency impairs cardiac interfibrillar mitochondrial function and myocardial contractility while inducing oxidative stress. Front Endocrinol (Lausanne) 2023; 14:1206387. [PMID: 37780627 PMCID: PMC10534000 DOI: 10.3389/fendo.2023.1206387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/06/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Clinical studies have shown that low levels of endogenous testosterone are associated with cardiovascular diseases. Considering the intimate connection between oxidative metabolism and myocardial contractility, we determined the effects of testosterone deficiency on the two spatially distinct subpopulations of cardiac mitochondria, subsarcolemmal (SSM) and interfibrillar (IFM). Methods We assessed cardiac function and cardiac mitochondria structure of SSM and IFM after 12 weeks of testosterone deficiency in male Wistar rats. Results and Discussion Results show that low testosterone reduced myocardial contractility. Orchidectomy increased total left ventricular mitochondrial protein in the SSM, but not in IFM. The membrane potential, size and internal complexity in the IFM after orchidectomy were higher compared to the SHAM group. However, the rate of oxidative phosphorylation with all substrates in the IFM after orchidectomy was lower compared to the SHAM group. Testosterone replacement restored these changes. In the testosterone-deficient SSM group, oxidative phosphorylation was decreased with palmitoyl-L-carnitine as substrate; however, the mitochondrial calcium retention capacity in IFM was increased. There was no difference in swelling of the mitochondria in either group. These changes in IFM were followed by a reduction in phosphorylated form of AMP-activated protein kinase (p-AMPK-α), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) translocation to mitochondria and decreased mitochondrial transcription factor A (TFAM). Testosterone deficiency increased NADPH oxidase (NOX), angiotensin converting enzyme (ACE) protein expression and reduced mitochondrial antioxidant proteins such as manganese superoxide dismutase (Mn-SOD) and catalase in the IFM. Treatment with apocynin (1.5 mM in drinking water) normalized myocardial contractility and interfibrillar mitochondrial function in the testosterone depleted animals. In conclusion, our findings demonstrate that testosterone deficiency leads to reduced myocardial contractility and impaired cardiac interfibrillar mitochondrial function. Our data suggest the involvement of reactive oxygen species, with a possibility of NOX as an enzymatic source.
Collapse
Affiliation(s)
| | - Karoline Sousa Ronconi
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Elis Aguiar Morra
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Paula Lopes Rodrigues
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Renata Andrade Ávila
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Merlo
- Department of Morphology, Federal University of Espírito Santo, Vitoria, ES, Brazil
| | - Jones B. Graceli
- Department of Morphology, Federal University of Espírito Santo, Vitoria, ES, Brazil
| | - Maylla Ronacher Simões
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Ivanita Stefanon
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | |
Collapse
|
14
|
Laasmaa M, Branovets J, Stolova J, Shen X, Rätsepso T, Balodis MJ, Grahv C, Hendrikson E, Louch WE, Birkedal R, Vendelin M. Cardiomyocytes from female compared to male mice have larger ryanodine receptor clusters and higher calcium spark frequency. J Physiol 2023; 601:4033-4052. [PMID: 37561554 DOI: 10.1113/jp284515] [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: 02/08/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023] Open
Abstract
Sex differences in cardiac physiology are receiving increased attention as it has become clear that men and women have different aetiologies of cardiac disease and require different treatments. There are experimental data suggesting that male cardiomyocytes exhibit larger Ca2+ transients due to larger Ca2+ sparks and a higher excitation-contraction coupling gain; in addition, they exhibit a larger response to adrenergic stimulation with isoprenaline (ISO). Here, we studied whether there are sex differences relating to structural organization of the transverse tubular network and ryanodine receptors (RyRs). Surprisingly, we found that female cardiomyocytes exhibited a higher spark frequency in a range of spark magnitudes. While overall RyR expression and phosphorylation were the same, female cardiomyocytes had larger but fewer RyR clusters. The density of transverse t-tubules was the same, but male cardiomyocytes had more longitudinal t-tubules. The Ca2+ transients were similar in male and female cardiomyocytes under control conditions and in the presence of ISO. The synchrony of the Ca2+ transients was similar between sexes as well. Overall, our data suggest subtle sex differences in the Ca2+ influx and efflux pathways and their response to ISO, but these differences are balanced, resulting in similar Ca2+ transients in field-stimulated male and female cardiomyocytes. The higher spark frequency in female cardiomyocytes is related to the organization of RyRs into larger, but fewer clusters. KEY POINTS: During a heartbeat, the force of contraction depends on the amplitude of the calcium transient, which in turn depends on the amount of calcium released as calcium sparks through ryanodine receptors in the sarcoplasmic reticulum. Previous studies suggest that cardiomyocytes from male compared to female mice exhibit larger calcium sparks, larger sarcoplasmic reticulum calcium release and greater response to adrenergic stimulation triggering a fight-or-flight response. In contrast, we show that cardiomyocytes from female mice have a higher spark frequency during adrenergic stimulation and similar spark morphology. The higher spark frequency is related to the organization of ryanodine receptors into fewer, but larger clusters in female compared to male mouse cardiomyocytes. Despite subtle sex differences in cardiomyocyte structure and calcium fluxes, the differences are balanced, leading to similar calcium transients in cardiomyocytes from male and female mice.
Collapse
Affiliation(s)
- Martin Laasmaa
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
- Institute for Experimental Medical Research, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Jekaterina Stolova
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Xin Shen
- Institute for Experimental Medical Research, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Triinu Rätsepso
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Mihkel Jaan Balodis
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Cärolin Grahv
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Eliise Hendrikson
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - William Edward Louch
- Institute for Experimental Medical Research, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
15
|
Raj A, Chakole S, Agrawal S, Gupta A, Khekade H, Prasad R, Lohakare T, Wanjari M. The Impact of Menopause on Cardiovascular Aging: A Comprehensive Review of Androgen Influences. Cureus 2023; 15:e43569. [PMID: 37719547 PMCID: PMC10503403 DOI: 10.7759/cureus.43569] [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: 07/03/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Menopause represents a critical life stage in women, characterized by hormonal changes that significantly impact cardiovascular health. While the decline in estrogen levels has long been recognized as a major contributor to cardiovascular aging in menopausal women, the role of androgens, particularly testosterone, has gained increasing attention in recent years. This comprehensive review aims to provide a thorough understanding of the impact of menopause on cardiovascular aging, with a specific focus on the influences of androgens. A literature search was conducted to gather relevant studies and clinical evidence exploring the relationship between menopause, androgens, and cardiovascular health. The review integrates findings from various studies to present a holistic view of the topic. The review outlines the changes in hormone levels during menopause and discusses the cardiovascular risk factors associated with this transition. Furthermore, it explores the impact of menopause on cardiovascular structure and function, elucidating the underlying mechanisms that contribute to cardiovascular aging. Androgens' significance in maintaining cardiovascular homeostasis is discussed, followed by exploring the effects of androgen decline during menopause on lipid profiles, insulin sensitivity, vascular function, and other cardiovascular parameters. The review delves into the mechanisms of androgen action on the cardiovascular system, emphasizing the role of androgen receptors and the intricate interplay between androgens, estrogens, and other hormones. Clinical evidence supporting the effects of androgens on cardiovascular aging is presented, including studies investigating the association between androgen levels and cardiovascular outcomes. Additionally, the impact of androgen replacement therapy (ART) on cardiovascular risk markers and events in menopausal women is examined, along with controversies and conflicting findings surrounding the use of androgen therapy in cardiovascular aging. This structured review provides a comprehensive understanding of the impact of menopause on cardiovascular aging, with a specific focus on the role of androgens. By highlighting the significance of androgens in cardiovascular health during menopause, this review aims to create an initial impression and interest among readers, inviting potential citations in the future. The findings underscore the need for further research and offer insights into managing cardiovascular aging in menopausal women, including lifestyle interventions, pharmacological approaches, and the potential role of personalized medicine and precision therapies.
Collapse
Affiliation(s)
- Aditya Raj
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Swarupa Chakole
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Suyash Agrawal
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anannya Gupta
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Harshal Khekade
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roshan Prasad
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tejaswee Lohakare
- Pediatrics, Smt. Radhikabai Meghe Memorial College of Nursing, Wardha, IND
| | - Mayur Wanjari
- Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
16
|
Edaravone alleviates methotrexate-induced testicular injury in rats: Implications on inflammation, steroidogenesis, and Akt/p53 signaling. Int Immunopharmacol 2023; 117:109969. [PMID: 37012866 DOI: 10.1016/j.intimp.2023.109969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023]
Abstract
Edaravone (ED) is a neuroprotective drug with beneficial effects against several disorders due to its prominent antioxidant activity. However, its effect against methotrexate (MTX)-induced testicular damage was not previously investigated. Therefore, we aimed to investigate the ability of ED to prevent the oxidative stress, inflammation, and apoptosis induced by MTX on the rat testis and to examine whether ED administration modulated the Akt/p53 signaling and steroidogenesis process. Rats were allocated into; Normal, ED (20 mg/kg, PO, for 10 days), MTX (20 mg/kg, i.p., on the 5th day), and ED + MTX groups. The results showed that MTX group exhibited higher serum activities of ALT, AST, ALP, and LDH in addition to histopathological alterations in the rat testis, compared to normal group. Furthermore, MTX induced down-regulation of the steroidogenic genes; StAR, CYP11a1, and HSD17B3 and reduced FSH, LH, and testosterone levels. The MTX group also showed higher levels of MDA, NO, MPO, NF-kB, TNF-α, IL-6, IL-1β, Bax, and caspase 3, as well as, lower levels of GSH, GPx, SOD, IL-10, Bcl2 compared to normal rats, p < 0.05. In addition, MTX treatment resulted in increased p53 expression and decreased p-Akt expression. Remarkably, ED administration significantly prevented all the biochemical, genetic, and histological damage induced by MTX. Hence, ED treatment protected the rat testis from apoptosis, oxidative stress, inflammation, and impaired steroidogenesis induced by MTX. This novel protective effect was mediated by decreasing p53 while increasing p-Akt protein expression.
Collapse
|
17
|
Alam N, Najnin H, Islam M, Shakya S, Khan IM, Zaidi R. Biochemical and histopathological analysis after sub-chronic administration of oxyresveratrol in Wistar rats. Drug Chem Toxicol 2023; 46:166-175. [PMID: 34913788 DOI: 10.1080/01480545.2021.2015243] [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: 02/02/2023]
Abstract
Oxyresveratrol (OXY) is a naturally occurring phenolic compound; however, there are no toxicity studies reported on its long term use. The aim of our work was to demonstrate the evaluation of acute and sub-chronic toxicity of oxyresveratrol in rats to assess its safety profile. To evaluate the LD50 value, 2000 mg/kg of oxyresveratrol was administered to Wistar rats by oral gavage. For sub-chronic toxicity assessment, 80 Wistar rats were randomly divided into four groups (10 animal/sex/group) and oxyresveratrol administered at a dose of 50, 100, 150 mg/kg/day by oral gavage. Bodyweight, food, and water consumption were monitored every week. At the end of the experiments, biochemical and hematological parameters were analyzed. Gross and microscopic organ analyses were also carried out. LD50 of oxyresveratrol was greater than 2000 mg/kg sub-chronic administration of oxyresveratrol did not influence any mortality. Doses of 50 and 100 mg/kg of oxyresveratrol did not produce any sign of toxicity. However, the 150 mg/kg oxyresveratrol group depicted changes in multiple biochemical and hematological parameters with changes in the pathology of cardiac, hepatic, and renal tissues when compared with control. Therefore, no observed adverse effect level (NOAEL) of oxyresveratrol was observed to be 100 mg/kg per day for both male and female rats.
Collapse
Affiliation(s)
- Nisat Alam
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Hasina Najnin
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Maidul Islam
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Sonam Shakya
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Ishaat M Khan
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Rana Zaidi
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
18
|
Hamam A, Abou-Omar M, Rabah H, Khattab H, Alaarag A. Worsening effect of testosterone deficiency on males with heart failure with preserved ejection fraction. BMC Endocr Disord 2022; 22:321. [PMID: 36529735 PMCID: PMC9761962 DOI: 10.1186/s12902-022-01249-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF)is challenging. Patients usually have normal LV size and ejection fraction. This clinical syndrome develops from a complex interaction of several risk factors that cause organ dysfunction and clinical symptoms. There's evidence that testosterone deficiency is associated with a worse cardiometabolic profile and increased inflammatory markers. We thought that these changes might have an impact on heart failure pathogenesis. We aimed to study the relationship between testosterone level and symptoms in HFpEF. METHODS We studied 120 male patients with HFpEF. According to New York Heart Association (NYHA), patients were classified into I, II and III classes; class IV patients were excluded. All patients were subjected to clinical and echocardiographic examinations. In addition, we measured serum testosterone, cardio-metabolic profile, intracellular adhesive molecule-1(ICAM-1), P-selectin and nitric oxide (NO) levels. RESULTS Patients with testosterone deficiency had worse NYHA class and higher BNP P = (0.001). Additionally, they had a significantly worse metabolic profile; higher total cholesterol, triglycerides, LDL cholesterol, fasting insulin and HOMA-IR P = (0.005, 0.001, 0.001, 0.001), respectively. Also, they had higher inflammatory markers and worse endothelial functional parameters; (ICAM-1, NO and P- selectin) P = (0.001). Age, BNP and testosterone deficiency can be used as independent predictors of NYHA class III symptoms with a Testosterone cutoff value of 2.7 ng/ml. CONCLUSION Testosterone deficiency could be used as an independent predictor of symptom severity in HFpEF, and it aggravates systemic inflammation and endothelial dysfunction in these patients.
Collapse
Affiliation(s)
- Ahmed Hamam
- Department of internal medicine, Armed Forces College of Medicine, Cairo, Egypt
| | - Mahmoud Abou-Omar
- Cardiovascular medicine department, Faculty of Medicine, Tanta University, Tanta, 31511, Egypt
| | - Hanem Rabah
- Faculty of Medicine, Department of medical biochemistry, Tanta University, Tanta, Egypt
| | - Haidy Khattab
- Faculty of Medicine Department of medical physiology, Tanta University, Tanta, Egypt
| | - Ahmed Alaarag
- Cardiovascular medicine department, Faculty of Medicine, Tanta University, Tanta, 31511, Egypt.
| |
Collapse
|
19
|
Njoroge JN, Tressel W, Biggs ML, Matsumoto AM, Smith NL, Rosenberg E, Hirsch CH, Gottdiener JS, Mukamal KJ, Kizer JR. Circulating Androgen Concentrations and Risk of Incident Heart Failure in Older Men: The Cardiovascular Health Study. J Am Heart Assoc 2022; 11:e026953. [PMID: 36285783 PMCID: PMC9673636 DOI: 10.1161/jaha.122.026953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Circulating androgen concentrations in men decline with age and have been linked to diabetes and atherosclerotic cardiovascular disease (ASCVD). A similar relationship has been reported for low total testosterone and incident heart failure (HF) but remains unstudied for free testosterone or the more potent androgen dihydrotestosterone (DHT). We hypothesized that total/free testosterone are inversely related, sex hormone–binding globulin is positively related, and total/free DHT bear a U‐shaped relationship with incident HF. Methods and Results In a sample of men from the CHS (Cardiovascular Health Study) without atherosclerotic cardiovascular disease or HF, serum testosterone and DHT concentrations were measured by liquid chromatography–tandem mass spectrometry, and sex hormone–binding globulin by immunoassay. Free testosterone or DHT was calculated from total testosterone or total DHT, sex hormone–binding globulin, and albumin. We used Cox regression to estimate relative risks of HF after adjustment for potential confounders. In 1061 men (aged 76±5 years) followed for a median of 9.6 years, there were 368 HF events. After adjustment, lower calculated free testosterone was significantly associated with higher risk of HF (hazard ratio [HR], 1.14 [95% CI, 1.01–1.28]). Risk estimates for total testosterone (HR, 1.12 [95% CI, 0.99–1.26]), total DHT (HR, 1.10 [95% CI, 0.97–1.24]), calculated free dihydrotestosterone (HR, 1.09 [95% CI, 0.97–1.23]), and sex hormone–binding globulin (HR, 1.07 [95% CI, 0.95–1.21]) were directionally similar but not statistically significant. Conclusions Calculated free testosterone was inversely associated with incident HF, suggesting a contribution of testosterone deficiency to HF incidence among older men. Additional research is necessary to determine whether testosterone replacement therapy might be an effective strategy to lower HF risk in older men.
Collapse
Affiliation(s)
| | | | | | - Alvin M. Matsumoto
- University of Washington Seattle WA
- Veterans Affairs Puget Sound Health Care System Seattle WA
| | | | - Emily Rosenberg
- Brigham and Women’s Hospital Boston MA
- Harvard Medical School Boston MA
| | | | | | - Kenneth J. Mukamal
- Harvard Medical School Boston MA
- Beth Israel Deaconess Medical Center Boston MA
| | - Jorge R. Kizer
- University of California San Francisco San Francisco CA
- San Francisco Veterans Affairs Health Care System San Francisco CA
| |
Collapse
|
20
|
Analyzing the Systems Biology Effects of COVID-19 mRNA Vaccines to Assess Their Safety and Putative Side Effects. Pathogens 2022; 11:pathogens11070743. [PMID: 35889989 PMCID: PMC9320269 DOI: 10.3390/pathogens11070743] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 01/25/2023] Open
Abstract
COVID-19 vaccines have been instrumental tools in reducing the impact of SARS-CoV-2 infections around the world by preventing 80% to 90% of hospitalizations and deaths from reinfection, in addition to preventing 40% to 65% of symptomatic illnesses. However, the simultaneous large-scale vaccination of the global population will indubitably unveil heterogeneity in immune responses as well as in the propensity to developing post-vaccine adverse events, especially in vulnerable individuals. Herein, we applied a systems biology workflow, integrating vaccine transcriptional signatures with chemogenomics, to study the pharmacological effects of mRNA vaccines. First, we derived transcriptional signatures and predicted their biological effects using pathway enrichment and network approaches. Second, we queried the Connectivity Map (CMap) to prioritize adverse events hypotheses. Finally, we accepted higher-confidence hypotheses that have been predicted by independent approaches. Our results reveal that the mRNA-based BNT162b2 vaccine affects immune response pathways related to interferon and cytokine signaling, which should lead to vaccine success, but may also result in some adverse events. Our results emphasize the effects of BNT162b2 on calcium homeostasis, which could be contributing to some frequently encountered adverse events related to mRNA vaccines. Notably, cardiac side effects were signaled in the CMap query results. In summary, our approach has identified mechanisms underlying both the expected protective effects of vaccination as well as possible post-vaccine adverse effects. Our study illustrates the power of systems biology approaches in improving our understanding of the comprehensive biological response to vaccination against COVID-19.
Collapse
|
21
|
King DR, Hardin KM, Hoeker GS, Poelzing S. Re-evaluating methods reporting practices to improve reproducibility: an analysis of methodological rigor for the Langendorff whole-heart technique. Am J Physiol Heart Circ Physiol 2022; 323:H363-H377. [PMID: 35749719 PMCID: PMC9359653 DOI: 10.1152/ajpheart.00164.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In recent decades, the scientific community has seen an increased interest in rigor and reproducibility. In 2017, concerns of methodological thoroughness and reporting practices were implicated as significant barriers to reproducibility within the preclinical cardiovascular literature, particularly in studies employing animal research. The Langendorff, whole-heart technique has proven to be an invaluable research tool, being modified in a myriad of ways to probe questions across the spectrum of physio- and pathophysiologic function of the heart. As a result, significant variability in the application of the Langendorff technique exists. This literature review quantifies the different methods employed in the implementation of the Langendorff technique and provides brief examples of how individual parametric differences can impact the outcomes and interpretation of studies. From 2017-2020, significant variability of animal models, anesthesia, cannulation time, and perfusate composition, pH, and temperature demonstrate that the technique has diversified to meet new challenges and answer different scientific questions. The review also reveals which individual methods are most frequently reported, even if there is no explicit agreement upon which parameters should be reported. The analysis of methods related to the Langendorff technique suggests a framework for considering methodological approach when interpreting seemingly contradictory results, rather than concluding that results are irreproducible.
Collapse
Affiliation(s)
- D Ryan King
- Translational Biology, Medicine, and Health Graduate Program. Virginia Polytechnic Institute and State University. Blacksburg, Virginia.,Dorothy M. Davis Heart and Lunch Research Institute, College of Medicine, The Ohio State University Wexner Medical Center. Columbus, Ohio
| | - Kathryn M Hardin
- Virginia Tech Carilion School of Medicine. Roanoke, Virginia.,Center for Heart and Reparative Medicine Research. Fralin Biomedical Research Institute at Virginia Tech Carilion. Roanoke, Virginia
| | - Gregory S Hoeker
- Center for Heart and Reparative Medicine Research. Fralin Biomedical Research Institute at Virginia Tech Carilion. Roanoke, Virginia
| | - Steven Poelzing
- Virginia Tech Carilion School of Medicine. Roanoke, Virginia.,Center for Heart and Reparative Medicine Research. Fralin Biomedical Research Institute at Virginia Tech Carilion. Roanoke, Virginia.,Department of Biomedical Engineering and Mechanics. Virginia Polytechnic Institute and State University. Blacksburg, Virginia
| |
Collapse
|
22
|
Usende IL, Oyelowo FO, Adikpe AO, Emikpe BO, Nafady AAHM, Olopade JO. Reproductive Hormones Imbalance, Germ Cell Apoptosis, Abnormal Sperm Morphophenotypes and Ultrastructural Changes in Testis of African Giant Rats (Cricetomys gambianus) Exposed to Sodium Metavanadate Intoxication. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:42849-42861. [PMID: 35088257 DOI: 10.1007/s11356-021-18246-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/16/2021] [Indexed: 06/14/2023]
Abstract
Environmental exposure to vanadium has been on the increase in recent time. This metal is a known toxicant. The current study was conducted to investigate the reproductive toxicity of sodium metavanadate (SMV) in male African giant rats. Administration of SMV was done intraperitoneally daily for 14 consecutive days at a dosage of 3 mg/kg body weight. Sterile water was administered to the control group. Serum reproductive hormones, sperm reserve and quality as well as testicular ultrastructural changes following SMV treatment were analysed. Results showed SMV-exposed AGR group had statistically decreased concentrations of testosterone (4.7 ng/ml), FSH (3.4 IU/L) and LH (3.8 IU/L). Also, SMV-treated group had statistically decreased sperm motility and mass activity with increased percentage of abnormal morphophenotypes of spermatozoa and upregulation of P53 immunopositive cells. Ultrastructural study revealed vacuolation of germ and Sertoli cells cytoplasm and nucleus, and mitochondrial swelling and vacuolations were also observed. There was severe disintegration of the seminiferous tubules, atrophy and degeneration of myeloid cells and apoptosis of the Leydig, Sertoli and germ cells. In conclusion, intraperitoneal SMV exposure exerts severe adverse effects on some serum reproductive hormones, reduction in the sperm reserve and quality, apoptosis and degenerative changes of the Leydig, Sertoli and germ cells which can lead to infertility.
Collapse
Affiliation(s)
- Ifukibot Levi Usende
- Department of Veterinary Anatomy, University of Abuja, Abuja, Nigeria.
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria.
| | | | - Agbonu Oluwa Adikpe
- Department of Veterinary Physiology and Biochemistry, University of Abuja, Abuja, Nigeria
| | | | | | | |
Collapse
|
23
|
Yusifov A, Woulfe KC, Bruns DR. Mechanisms and implications of sex differences in cardiac aging. THE JOURNAL OF CARDIOVASCULAR AGING 2022; 2:20. [PMID: 35419571 PMCID: PMC9004711 DOI: 10.20517/jca.2022.01] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Aging promotes structural and functional remodeling of the heart, even in the absence of external factors. There is growing clinical and experimental evidence supporting the existence of sex-specific patterns of cardiac aging, and in some cases, these sex differences emerge early in life. Despite efforts to identify sex-specific differences in cardiac aging, understanding how these differences are established and regulated remains limited. In addition to contributing to sex differences in age-related heart disease, sex differences also appear to underlie differential responses to cardiac stress such as adrenergic activation. Identifying the underlying mechanisms of sex-specific differences may facilitate the characterization of underlying heart disease phenotypes, with the ultimate goal of utilizing sex-specific therapeutic approaches for cardiac disease. The purpose of this review is to discuss the mechanisms and implications of sex-specific cardiac aging, how these changes render the heart more susceptible to disease, and how we can target age- and sex-specific differences to advance therapies for both male and female patients.
Collapse
Affiliation(s)
- Aykhan Yusifov
- Kinesiology & Health, University of Wyoming, Laramie, WY 82071, USA
| | - Kathleen C. Woulfe
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Danielle R. Bruns
- Kinesiology & Health, University of Wyoming, Laramie, WY 82071, USA
- Wyoming WWAMI Medical Education, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
24
|
Could Lower Testosterone in Older Men Explain Higher COVID-19 Morbidity and Mortalities? Int J Mol Sci 2022; 23:ijms23020935. [PMID: 35055119 PMCID: PMC8781054 DOI: 10.3390/ijms23020935] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 01/08/2023] Open
Abstract
The health scourge imposed on humanity by the COVID-19 pandemic seems not to recede. This fact warrants refined and novel ideas analyzing different aspects of the illness. One such aspect is related to the observation that most COVID-19 casualties were older males, a tendency also noticed in the epidemics of SARS-CoV in 2003 and the Middle East respiratory syndrome in 2012. This gender-related difference in the COVID-19 death toll might be directly involved with testosterone (TEST) and its plasmatic concentration in men. TEST has been demonstrated to provide men with anti-inflammatory and immunological advantages. As the plasmatic concentration of this androgen decreases with age, the health benefit it confers also diminishes. Low plasmatic levels of TEST can be determinant in the infection’s outcome and might be related to a dysfunctional cell Ca2+ homeostasis. Not only does TEST modulate the activity of diverse proteins that regulate cellular calcium concentrations, but these proteins have also been proven to be necessary for the replication of many viruses. Therefore, we discuss herein how TEST regulates different Ca2+-handling proteins in healthy tissues and propose how low TEST concentrations might facilitate the replication of the SARS-CoV-2 virus through the lack of modulation of the mechanisms that regulate intracellular Ca2+ concentrations.
Collapse
|
25
|
Ferreira C, Trindade F, Ferreira R, Neves JS, Leite-Moreira A, Amado F, Santos M, Nogueira-Ferreira R. Sexual dimorphism in cardiac remodeling: the molecular mechanisms ruled by sex hormones in the heart. J Mol Med (Berl) 2021; 100:245-267. [PMID: 34811581 DOI: 10.1007/s00109-021-02169-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/16/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is growing in prevalence, due to an increase in aging and comorbidities. Heart failure with reduced ejection fraction (HFrEF) is more common in men, whereas heart failure with preserved ejection fraction (HFpEF) has a higher prevalence in women. However, the reasons for these epidemiological trends are not clear yet. Since HFpEF affects mostly postmenopausal women, sex hormones should play a pivotal role in HFpEF development. Furthermore, for HFpEF, contrary to HFrEF, effective therapeutic approaches are missing. Interestingly, studies evidenced that some therapies can have better results in women than in HFpEF men, emphasizing the necessity of understanding these observations at a molecular level. Thus, herein, we review the molecular mechanisms of estrogen and androgen actions in the heart in physiological conditions and explain how its dysregulation can lead to disease development. This clarification is essential in the road for an effective personalized management of HF, particularly HFpEF, towards the development of sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Cláudia Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Fábio Trindade
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - João Sérgio Neves
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mário Santos
- Department of Cardiology, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rita Nogueira-Ferreira
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal.
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
26
|
Mitogen-activated protein kinases contribute to temperature-induced cardiac remodelling in rainbow trout (Oncorhynchus mykiss). J Comp Physiol B 2021; 192:61-76. [PMID: 34586481 DOI: 10.1007/s00360-021-01406-5] [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: 06/25/2021] [Revised: 08/16/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Rainbow trout (Oncorhynchus mykiss) live in environments where water temperatures range between 4 °C and 20 °C. Laboratory studies demonstrate that cold and warm acclimations of male trout can have oppositional effects on cardiac hypertrophy and the collagen content of the heart. The cellular mechanisms behind temperature-induced cardiac remodelling are unclear, as is why this response differs between male and female fish. Studies with cultured trout cardiac fibroblasts suggests that collagen deposition is regulated, at least in part, by mitogen-activated protein kinase (MAPK) cell signalling pathways. We, therefore, hypothesized that temperature-dependent cardiac remodelling is regulated by these signalling pathways. To test this, male and female trout were acclimated to 18 °C (warm) in the summer and to 4 °C (cold) in the winter and the activation of MAPK pathways in the hearts were characterized and compared to that of control fish maintained at 12 °C. In addition, cardiac collagen content, cardiac morphology and the expression of gene transcripts for matrix metalloproteinases (MMP) -9, MMP-2, tissue inhibitor of matrix metalloproteinases and collagen 1α were characterized. p38 MAPK phosphorylation increased in the hearts of female fish with cold acclimation and the phosphorylation of extracellular signal-regulated kinase increased in the hearts of male fish with warm acclimation. However, there was no effect of thermal acclimation on cardiac morphology or collagen content in either male or female fish. These results indicate that thermal acclimation has transient and sex-specific effects on the phosphorylation of MAPKs but also how variable the response of the trout heart is to thermal acclimation.
Collapse
|
27
|
Rapid Differentiation of Human Embryonic Stem Cells into Testosterone-Producing Leydig Cell-Like Cells In vitro. Tissue Eng Regen Med 2021; 18:651-662. [PMID: 34165777 PMCID: PMC8325741 DOI: 10.1007/s13770-021-00359-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Leydig cells (LCs) are testicular somatic cells that are the major producers of testosterone in males. Testosterone is essential for male physiology and reproduction. Reduced testosterone levels lead to hypogonadism and are associated with diverse pathologies, such as neuronal dysfunction, cardiovascular disease, and metabolic syndrome. LC transplantation is a promising therapy for hypogonadism; however, the number of LCs in the testis is very rare and they do not proliferate in vitro. Therefore, there is a need for an alternative source of LCs. Methods: To develop a safer, simple, and rapid strategy to generate human LC-like cells (LLCs) from stem cells, we first performed preliminary tests under different conditions for the induction of LLCs from human CD34/CD73 double positive-testis-derived stem cells (HTSCs). Based on the embryological sequence of events, we suggested a 3-step strategy for the differentiation of human ESCs into LLCs. We generated the mesendoderm in the first stage and intermediate mesoderm (IM) in the second stage and optimized the conditions for differentiation of IM into LLCs by comparing the secreted testosterone levels of each group. Results: HTSCs and human embryonic stem cells can be directly differentiated into LLCs by defined molecular compounds within a short period. Human ESC-derived LLCs can secrete testosterone and express steroidogenic markers. Conclusion: We developed a rapid and efficient protocol for the production of LLCs from stem cells using defined molecular compounds. These findings provide a new therapeutic cell source for male hypogonadism.
Collapse
|
28
|
Gasperetti A, James CA, Cerrone M, Delmar M, Calkins H, Duru F. Arrhythmogenic right ventricular cardiomyopathy and sports activity: from molecular pathways in diseased hearts to new insights into the athletic heart mimicry. Eur Heart J 2021; 42:1231-1243. [PMID: 33200174 DOI: 10.1093/eurheartj/ehaa821] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/12/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited disease associated with a high risk of sudden cardiac death. Among other factors, physical exercise has been clearly identified as a strong determinant of phenotypic expression of the disease, arrhythmia risk, and disease progression. Because of this, current guidelines advise that individuals with ARVC should not participate in competitive or frequent high-intensity endurance exercise. Exercise-induced electrical and morphological para-physiological remodelling (the so-called 'athlete's heart') may mimic several of the classic features of ARVC. Therefore, the current International Task Force Criteria for disease diagnosis may not perform as well in athletes. Clear adjudication between the two conditions is often a real challenge, with false positives, that may lead to unnecessary treatments, and false negatives, which may leave patients unprotected, both of which are equally inacceptable. This review aims to summarize the molecular interactions caused by physical activity in inducing cardiac structural alterations, and the impact of sports on arrhythmia occurrence and other clinical consequences in patients with ARVC, and help the physicians in setting the two conditions apart.
Collapse
Affiliation(s)
- Alessio Gasperetti
- Division of Cardiology, University Heart Center Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | - Cynthia A James
- Division of Cardiology, Johns Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Marina Cerrone
- Leon H Charney Division of Cardiology, New York University School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Mario Delmar
- Leon H Charney Division of Cardiology, New York University School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Hugh Calkins
- Division of Cardiology, Johns Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Firat Duru
- Division of Cardiology, University Heart Center Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Rämistrasse 71, Zurich 8006, Switzerland
| |
Collapse
|
29
|
Salama NAS, Salama G. Do sex hormones impact stress responses by modulating the cellular composition of the myocardium? Cardiovasc Res 2021; 117:2140-2142. [PMID: 33693660 DOI: 10.1093/cvr/cvab072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Noah A S Salama
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, 14642
| | - Guy Salama
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261
| |
Collapse
|
30
|
Keen J, Prisco SZ, Prins KW. Sex Differences in Right Ventricular Dysfunction: Insights From the Bench to Bedside. Front Physiol 2021; 11:623129. [PMID: 33536939 PMCID: PMC7848185 DOI: 10.3389/fphys.2020.623129] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/04/2022] Open
Abstract
There are inherent distinctions in right ventricular (RV) performance based on sex as females have better RV function than males. These differences are magnified and have very important prognostic implications in two RV-centric diseases, pulmonary hypertension (PH), and arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D). In both PH and ARVC/D, RV dysfunction results in poor patient outcomes. However, there are no currently approved therapies specifically targeting the failing RV, an important unmet need for these two life-threatening disorders. In this review, we highlight human data demonstrating divergent RV phenotypes in healthy, PH, and ARVC/D patients based on sex. Furthermore, we discuss the links between estrogen (the female predominant sex hormone), testosterone (the male predominant sex hormone), and dehydroepiandrosterone (a precursor hormone for multiple sex hormones in males and females) and RV function in both disorders. To provide potential mechanistic insights into sex differences in RV function, we review data that investigate how sex hormones combat or contribute to pathophysiological changes in the RV. Finally, we highlight the ongoing clinical trials in pulmonary arterial hypertension targeting estrogen and dehydroepiandrosterone signaling. Hopefully, a greater understanding of the factors that promote superior RV function in females will lead to novel therapeutic approaches to combat RV dysfunction in PH and ARVC/D.
Collapse
Affiliation(s)
- Jennifer Keen
- Pulmonary and Critical Care, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Sasha Z Prisco
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Kurt W Prins
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
31
|
Matta MG, Gulayin PE, García-Zamora S, Gutierrez L, Rubinstein AL, Irazola VE, Poggio R, Gonzalez-Zuelgaray J. Epidemiology of early repolarization pattern in an adult general population. Acta Cardiol 2020; 75:713-723. [PMID: 31526309 DOI: 10.1080/00015385.2019.1667623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background: Early repolarization pattern (ERP) is a frequent finding in asymptomatic subjects with controversial implications regarding to its prognosis. This study aims to estimate the prevalence of ERP and its association with sociodemographic characteristics and cardiovascular risk factors among the adult population in the Southern Cone of Latin America.Methods: A sub-sample of 5398 participants of the CESCAS I study was included in the present analysis. ERP was defined as a J peak ≥0.1 mV in two or more contiguous leads with an end-QRS notch or slur on the downslope of a prominent R-wave.Results: The global prevalence of ERP was 8.1%; 11.1% in men and 5.6% in women. The prevalence in women increased with age (odds ratio [OR] 2.5, 95% confidence interval [CI] 1.5-4.2, at >65 years, p < 0.001), current cigarette smoking (OR 1.4, 95%CI 1.0-2.0, p = 0.045) and hypercholesterolaemia (OR 1.4, 95%CI 1.0-2.0, 0 p = 0.036). Conversely, in men, ERP prevalence decreased with age (OR 0.5, 95%CI 0.3-0.9, at >65 years, p = 0.01) and obesity (OR 0.6, 95%CI 0.4-0.8, p = 0.006). We found an increasing ERP prevalence with a higher Sokolow-Lyon index in both sexes (p < 0.001). Inferior location was found in 67.9% of cases, and the most common ERP type was a "slurring" appearance without ST elevation (76.3%).Conclusions: We found an overall prevalence of ERP of 8.1% and a robust association of ERP with normal BMI and higher Sokolow-Lyon index in men and with hypercholesterolaemia, current cigarette smoking and higher Sokolow-Lyon index in women.
Collapse
Affiliation(s)
- María Gabriela Matta
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Pablo Elias Gulayin
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Sebastián García-Zamora
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Laura Gutierrez
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | | | - Vilma Edith Irazola
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Rosana Poggio
- South American Center of Excellence for Cardiovascular Health (CESCAS), Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Jorge Gonzalez-Zuelgaray
- Institute of Cardiological Research, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| |
Collapse
|
32
|
Abstract
PURPOSE OF THE REVIEW This review summarizes sex-related changes in the heart and vasculature that occur with aging, both in the presence and absence of cardiovascular disease (CVD). RECENT FINDINGS In the presence of CVD risk factors and/or overt CVD, sex-specific changes in the number of cardiomyocytes, extent of the myocardial extracellular matrix, and myocellular hypertrophy promote unique patterns of LV remodeling in men and women. In addition, age- and sex-specific vascular stiffening is also well established, driven by changes in endothelial dysfunction, elastin-collagen content, microvascular dysfunction, and neurohormonal signaling. Together, these changes in LV chamber geometry and morphology, coupled with heightened vascular stiffness, appear to drive both age-related increases in systolic function and declines in diastolic function, particularly in postmenopausal women. Accordingly, estrogen has been implicated as a key mediator, given its direct vasodilating properties, association with nitric oxide excretion, and involvement in myocellular Ca2+ handling, mitochondrial energy production, and oxidative stress. The culmination of the abovementioned sex-specific cardiac and vascular changes across the lifespan provides important insight into heart failure development, particularly of the preserved ejection fraction variety, while offering promise for future preventive strategies and therapeutic approaches.
Collapse
Affiliation(s)
- Andrew Oneglia
- Applied Physiology and Advanced Imaging Lab, University of Texas at Arlington, 655 West Mitchell St, Arlington, TX, 76010, USA
| | - Michael D Nelson
- Applied Physiology and Advanced Imaging Lab, University of Texas at Arlington, 655 West Mitchell St, Arlington, TX, 76010, USA
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vicente Blvd, AHSP Suite A3206, Los Angeles, CA, 90048, USA
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vicente Blvd, AHSP Suite A3206, Los Angeles, CA, 90048, USA.
| |
Collapse
|
33
|
Kalra S, Bhattacharya S, Kalhan A. Testosterone in COVID-19 - Foe, Friend or Fatal Victim? EUROPEAN ENDOCRINOLOGY 2020; 16:88-91. [PMID: 33117437 PMCID: PMC7572157 DOI: 10.17925/ee.2020.16.2.88] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023]
Abstract
The evidence derived from observational studies suggests male gender, diabetes and central obesity to be risk factors associated with an increased COVID-19-related case fatality. The precise pathophysiology behind this gender difference in mortality outcomes remains unclear at this stage, although it is worth exploring a possible role of testosterone as one of the contributory factors. The observed role of androgens in transcription of transmembrane protease serine-2, which facilitates COVID-19 anchoring to angiotensin-converting enzyme 2 cell surface receptors, seems to suggest that higher testosterone levels might be detrimental for outcomes. On the other hand, men with type 2 diabetes mellitus and central obesity have an increased prevalence of hypogonadotropic hypogonadism, with inhibition of gonadotropin-releasing hormone secretion induced by inflammatory cytokines being one of the postulated mechanisms. The increased COVID-19 case fatality in this cohort might perhaps reflect an underlying pro-inflammatory state, with low testosterone levels being either a surrogate marker of a poor metabolic state or playing a more active role in propagation of inflammation and thrombosis.
Collapse
Affiliation(s)
- Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
| | | | - Atul Kalhan
- Department of Endocrinology, Royal Glamorgan Hospital, Cardiff, UK
| |
Collapse
|
34
|
Ren J, Chen L, Zhang N, Chen X, Zhao Q, Chen K, Li X, Ruschitzka F, Duru F, Song J. Plasma testosterone and arrhythmic events in male patients with arrhythmogenic right ventricular cardiomyopathy. ESC Heart Fail 2020; 7:1547-1559. [PMID: 32469163 PMCID: PMC7373945 DOI: 10.1002/ehf2.12704] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/13/2020] [Accepted: 03/30/2020] [Indexed: 01/23/2023] Open
Abstract
AIMS Arrhythmogenic right ventricular cardiomyopathy (ARVC) is associated with life-threatening ventricular arrhythmia and progressive ventricular dysfunction. Previous studies suggested that sex hormones play an important role in the onset and prognosis of ARVC. This study aimed to investigate the role of testosterone in predicting major adverse cardiac events in the Chinese ARVC cohort. METHODS AND RESULTS Ninety-nine ARVC patients (median age, 40 years; 70.7% male) and 96 healthy controls (median age, 41 years; 62.5% male) were enrolled. The circulating levels of testosterone were measured by enzyme-linked immunosorbent assays (ELISA). The median follow-up time of all ARVC male patients was 17 months (interquartile range/IQR 9-29). Cox proportional hazards regression was used to analyse the effect of plasma testosterone and other well-described risk factors on malignant arrhythmic events in male ARVC patients. The male ARVC patients had significantly elevated levels of total testosterone [TT, 6.390 (4.438-8.768) ng/mL vs. 3.617 (2.073-4.479) ng/mL, P < 0.0001, data shown as the median with IQR], bioavailable testosterone [BT, 4.11 (1.990-6.545) ng/mL vs. 1.32 (0.7965-2.0350) ng/mL, P < 0.0001, median with IQR], and free testosterone [FT, 0.2055 (0.1000-0.4073) ng/mL vs. 0.0768 (0.0405-0.1105) ng/mL, P < 0.0001, median with IQR] than healthy male volunteer, whereas no differences were observed among female counterparts. There was no significant correlation between the baseline clinical characteristics and testosterone levels in male ARVC patients (Spearman's correlation test, P > 0.05). During the follow-up, the levels of testosterone were higher in male patients who experienced malignant arrhythmic events (N = 22) than in those who did not (N = 25) [TT, 9.034 (7.222-15.370) ng/mL vs. 4.633 (3.363-6.375) ng/mL, P < 0.001; BT, 7.485 (2.070-9.163) ng/mL vs. 3.300 (1.685-4.690) ng/mL, P < 0.05; FT, 0.453 (0.221-0.758) ng/mL vs. 0.161 (0.075-0.337) ng/mL P < 0.05, data expressed as median (IQR) and adjusted by Dunn's multiple comparisons test], whereas such distinction was not observed among patients with significant structural progression events (N = 16). Through multivariable adjustments, the Cox regression analysis showed the level of plasma total testosterone (HR = 1.325, 95% confidence interval = 1.171-1.498, P < 0.001) was an independent predictor for malignant arrhythmic events. CONCLUSIONS The levels of plasma testosterone in ARVC male patients are higher than those in healthy males. Testosterone level, without relation to the baseline cardiac function and future significant structural progression events, is a strong predictor of future adverse arrhythmic events in male patients with ARVC. Therefore, our results suggest that testosterone may be a useful biomarker in arrhythmic risk prediction in the ARVC.
Collapse
Affiliation(s)
- Jie Ren
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangjie Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Firat Duru
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, University Heart Center, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Maione AS, Pilato CA, Casella M, Gasperetti A, Stadiotti I, Pompilio G, Sommariva E. Fibrosis in Arrhythmogenic Cardiomyopathy: The Phantom Thread in the Fibro-Adipose Tissue. Front Physiol 2020; 11:279. [PMID: 32317983 PMCID: PMC7147329 DOI: 10.3389/fphys.2020.00279] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disorder, predisposing to malignant ventricular arrhythmias leading to sudden cardiac death, particularly in young and athletic patients. Pathological features include a progressive loss of myocardium with fibrous or fibro-fatty substitution. During the last few decades, different clinical aspects of ACM have been well investigated but still little is known about the molecular mechanisms that underlie ACM pathogenesis, leading to these phenotypes. In about 50% of ACM patients, a genetic mutation, predominantly in genes that encode for desmosomal proteins, has been identified. However, the mutation-associated mechanisms, causing the observed cardiac phenotype are not always clear. Until now, the attention has been principally focused on the study of molecular mechanisms that lead to a prominent myocardium adipose substitution, an uncommon marker for a cardiac disease, thus often recognized as hallmark of ACM. Nonetheless, based on Task Force Criteria for the diagnosis of ACM, cardiomyocytes death associated with fibrous replacement of the ventricular free wall must be considered the main tissue feature in ACM patients. For this reason, it urges to investigate ACM cardiac fibrosis. In this review, we give an overview on the cellular effectors, possible triggers, and molecular mechanisms that could be responsible for the ventricular fibrotic remodeling in ACM patients.
Collapse
Affiliation(s)
- Angela Serena Maione
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Chiara Assunta Pilato
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michela Casella
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Alessio Gasperetti
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
- University Heart Center, Zurich University Hospital, Zurich, Switzerland
| | - Ilaria Stadiotti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
36
|
KITTNAR O. Selected Sex Related Differences in Pathophysiology of Cardiovascular System. Physiol Res 2020; 69:21-31. [DOI: 10.33549/physiolres.934068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The annual incidence of cardiovascular diseases is age-dependently increasing both in men and women, however, the prevalence is higher in men until midlife. The higher incidence of cardiovascular disease in men than in women of similar age, and the menopause-associated increase in cardiovascular disease in women, has led to speculation that gender-related differences in sex hormones might have a key role in the development and evolution of cardiovascular disease. There are several suggested pathways in which gender and sex hormones can affect human cardiovascular system to produce original sexually different pathophysiology between women and men. Sex steroid hormones and their receptors are critical determinants of cardiovascular gender differences. Also arterial blood pressure is typically lower in women than in men what could be explained particularly by greater synthesis of nitric oxide (NO) in women. Female cardiomyocytes have a greater survival advantage when challenged with oxidative stress, suggesting that female hormones may play an important role in antioxidative protection of myocardium. It was also demonstrated in animal models that combination of XX chromosomes versus an XY chromosomes enhances sex differences in higher HDL cholesterol. Women were found to have reduced sympathetic activity (reflected by lower total peripheral resistance) and pulmonary artery pressure and enhanced parasympathetic activity relative to men. Similarly, men were found to have higher plasma norepinephrine levels than women. Regarding differences between the sexes in electrophysiology of the heart, two principle mechanisms have been proposed to explain them: hormonal effects on the expression or function of ion channels or, conversely, differences in autonomic tone. To improve diagnosis and treatment of cardiovascular diseases, greater focus on understanding the molecular and cellular physiology of the sex steroid hormones and their receptors in the cardiovascular system will be required.
Collapse
Affiliation(s)
- O. KITTNAR
- Institute of Physiology of the First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
37
|
Bruno C, Silvestrini A, Calarco R, Favuzzi AMR, Vergani E, Nicolazzi MA, d'Abate C, Meucci E, Mordente A, Landolfi R, Mancini A. Anabolic Hormones Deficiencies in Heart Failure With Preserved Ejection Fraction: Prevalence and Impact on Antioxidants Levels and Myocardial Dysfunction. Front Endocrinol (Lausanne) 2020; 11:281. [PMID: 32477267 PMCID: PMC7235369 DOI: 10.3389/fendo.2020.00281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose: In heart failure with reduced ejection fraction, catabolic mechanisms have a strong negative impact on mortality and morbidity. The relationship between anabolic hormonal deficiency and heart failure with preserved ejection fraction (HFpEF) has still been poorly investigated. On the other hand, oxidative stress is recognized as a player in the pathogenesis of HFpEF. Therefore, we performed a cohort study in HFpEF aimed to (1) define the multi-hormonal deficiency prevalence in HFpEF patients; (2) investigate the relationships between hormonal deficiencies and echocardiographic indexes; (3) explore the modulatory activity of anabolic hormones on antioxidant systems. Methods: 84 patients with diagnosis of HFpEF were enrolled in the study. Plasma levels of N-terminal pro-brain natriuretic peptide, fasting glucose, insulin, lipid pattern, insulin-like growth factor-1, dehydroepiandrosterone-sulfate (DHEA-S), total testosterone (T, only in male subjects) were evaluated. Hormonal deficiencies were defined according to T.O.S.C.A. multi-centric study, as previously published. An echocardiographic evaluation was performed. Plasma total antioxidant capacity (TAC) was measured using the system metmyoglobin -H2O2 and the chromogen ABTS, whose radical form is spectroscopically revealed; latency time (LAG) in the appearance of ABTS• is proportional to antioxidants in sample. Results: Multiple deficiencies were discovered. DHEA-S deficiency in 87% of patients, IGF-1 in 67% of patients, T in 42%. Patients with DHEA-S deficiency showed lower levels of TAC expressed by LAG (mean ± SEM 91.25 ± 9.34 vs. 75.22 ± 4.38 s; p < 0.05). No differences between TAC in patients with or without IGF-1 deficiency were found. A trend toward high level of TAC in patients without hormonal deficiencies compared with patients with one or multiple deficiencies was found. Regarding echocardiographic parameters, Left Atrial and Left Atrial Volume Index were significantly higher in patients with low IGF-1 values (mean ± SD 90.84 ± 3.86 vs. 72.83 ± 3.78 mL; 51.03 ± 2.33 vs. 40.56 ± 2.46 mL/m2, respectively; p < 0.05). Conclusions: Our study showed high prevalence of anabolic deficiencies in HFpEF. DHEA-S seems to influence antioxidant levels; IGF-1 deficiency was associated with alteration in parameters of myocardial structure and dysfunction. These data suggest a role of anabolic hormones in the complex pathophysiological mechanisms of HFpEF and could represent the basis for longitudinal studies and investigations on possible benefits of replacement therapy.
Collapse
Affiliation(s)
- Carmine Bruno
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Andrea Silvestrini
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- *Correspondence: Andrea Silvestrini
| | - Rodolfo Calarco
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Angela M. R. Favuzzi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Edoardo Vergani
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Maria Anna Nicolazzi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Claudia d'Abate
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Elisabetta Meucci
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alvaro Mordente
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Raffaele Landolfi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
| | - Antonio Mancini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario a Gemelli IRCCS, Rome, Italy
- Antonio Mancini
| |
Collapse
|
38
|
Su X, Lin D, Luo D, Sun M, Wang X, Ye J, Zhang M, Zhang Y, Xu X, Yu C, Guan Q. Cyclophilin D participates in the inhibitory effect of high-fat diet on the expression of steroidogenic acute regulatory protein. J Cell Mol Med 2019; 23:6859-6871. [PMID: 31373170 PMCID: PMC6787510 DOI: 10.1111/jcmm.14569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The high-fat diet (HFD)-induced obesity is responsible for the testosterone deficiency (TD). However, the mechanism remains unknown. Mitochondrial homeostasis is proved to be important for maintaining the function of steroidogenic acute regulatory protein (StAR), the first rate-limiting enzyme in testosterone synthesis. As the key regulator of mitochondrial membrane permeability, cyclophilin D (CypD) plays a crucial role in maintaining mitochondrial function. In this study, we sought to elucidate the role of CypD in the expression of StAR affected by HFD. METHODS To analyse the influence of CypD on StAR in vivo and in vitro, mouse models of HFD, CypD overexpression and CypD knockout (Ppif-/- ) as well as Leydig cells treated with palmitic acid (PA) and CypD overexpression plasmids were examined with an array of metabolic, mitochondrial function and molecular assays. RESULTS Compared with the normal diet mice, consistent with reduced testosterone in testes, the expressions of StAR in both mRNA and protein levels in HFD mice were down-regulated, while expressions of CypD were up-regulated. High-fat intake impaired mitochondrial function with the decrease in StAR in Leydig cells. Overexpression of CypD inhibited StAR expressions in vivo and in vitro. Compared with C57BL/6 mice with HFD, expressions of StAR were improved in Ppif-/- mice with HFD. CONCLUSIONS Mitochondrial CypD involved in the inhibitory effect of HFD on StAR expression in testes.
Collapse
Affiliation(s)
- Xiaohui Su
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
- General PracticeJinan City People's HospitalJinanChina
| | - Dong Lin
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
- Peking Union Medical College, Chinese Academy of Medical SciencesGraduate School of Peking Union Medical CollegeBeijingChina
| | - Dandan Luo
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Mingqi Sun
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Xiaolei Wang
- Shandong Institute of Endocrine & Metabolic DiseasesShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Jifeng Ye
- Department of Endocrinology and MetabolismThe Second People's Hospital of LiaochengLiaochengChina
| | - Meijie Zhang
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Yikun Zhang
- Shandong Institute of Endocrine & Metabolic DiseasesShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Xiaolin Xu
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
- Department of Rheumatology and ImmunologyDongying People's HospitalDongyingChina
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Key Laboratory of Endocrinology and Lipid MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| |
Collapse
|
39
|
Dibaba DT, Xun P, Fly AD, Bidulescu A, Tsinovoi CL, Judd SE, McClure LA, Cushman M, Unverzagt FW, He K. Calcium Intake and Serum Calcium Level in Relation to the Risk of Ischemic Stroke: Findings from the REGARDS Study. J Stroke 2019; 21:312-323. [PMID: 31590475 PMCID: PMC6780017 DOI: 10.5853/jos.2019.00542] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/28/2019] [Accepted: 08/27/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Data on the association between calcium (Ca) and ischemic stroke are sparse and inconsistent. This study aimed to examine Ca intake and serum Ca levels in relation to risk of ischemic stroke. METHODS The primary analysis included 19,553 participants from the Reasons for Geographic And Racial Differences in Stroke (REGARDS) study. A subcohort was randomly selected to create a case-cohort study (n=3,016), in which serum Ca levels were measured. Ischemic stroke cases were centrally adjudicated by physicians based on medical records. Cox proportional hazards regression for the cohort and weighted Cox proportional hazard regression with robust sandwich estimation method for the case-cohort analysis with adjustment for potential confounders were performed. RESULTS During a mean 8.3-year follow-up, 808 incident cases of ischemic stroke were documented. Comparing the highest quintile to the lowest, a statistically significant inverse association was observed between total Ca intake and risk of ischemic stroke (hazard ratio [HR], 0.72; 95% confidence interval [CI], 0.55 to 0.95; Plinear-trend=0.183); a restricted cubic spline analysis indicated a threshold effect like non-linear association of total Ca intake with ischemic stroke (Pnon-linear=0.006). In the case-cohort, serum Ca was inversely associated with the risk of ischemic stroke. Compared to the lowest, the highest quintile of serum Ca had a 27% lower risk of ischemic stroke (HR, 0.73; 95% CI, 0.53 to 0.99; Plinear-trend=0.013). Observed associations were mainly mediated by type 2 diabetes, hypertension, and cholesterol. CONCLUSION s These findings suggest that serum Ca has inverse and Ca intake has threshold effect like association with risk of ischemic stroke.
Collapse
Affiliation(s)
- Daniel T. Dibaba
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
- Department of Vice Chancellor for Research/Tennessee Clinical and Translational Science Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Pengcheng Xun
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
| | - Alyce D. Fly
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
| | - Aurelian Bidulescu
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
| | - Cari L. Tsinovoi
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
| | - Suzanne E. Judd
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leslie A. McClure
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA, USA
| | - Mary Cushman
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Ka He
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
40
|
Ayaz O, Banga S, Heinze-Milne S, Rose RA, Pyle WG, Howlett SE. Long-term testosterone deficiency modifies myofilament and calcium-handling proteins and promotes diastolic dysfunction in the aging mouse heart. Am J Physiol Heart Circ Physiol 2019; 316:H768-H780. [PMID: 30657724 DOI: 10.1152/ajpheart.00471.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The impact of long-term gonadectomy (GDX) on cardiac contractile function was explored in the setting of aging. Male mice were subjected to bilateral GDX or sham operation (4 wk) and investigated at 16-18 mo of age. Ventricular myocytes were field stimulated (2 Hz, 37°C). Peak Ca2+ transients (fura 2) and contractions were similar in GDX and sham-operated mice, although Ca2+ transients (50% decay time: 45.2 ± 2.3 vs. 55.6 ± 3.1 ms, P < 0.05) and contractions (time constant of relaxation: 39.1 ± 3.2 vs. 69.5 ± 9.3 ms, P < 0.05) were prolonged in GDX mice. Action potential duration was increased in myocytes from GDX mice, but this did not account for prolonged responses, as Ca2+ transient decay was slow even when cells from GDX mice were voltage clamped with simulated "sham" action potentials. Western blots of proteins involved in Ca2+ sequestration and efflux showed that Na+/Ca2+ exchanger and sarco(endo)plasmic reticulum Ca2+-ATPase type 2 protein levels were unaffected, whereas phospholamban was dramatically higher in ventricles from aging GDX mice (0.24 ± 0.02 vs. 0.86 ± 0.13, P < 0.05). Myofilament Ca2+ sensitivity at physiological Ca2+ was similar, but phosphorylation of essential myosin light chain 1 was reduced by ≈50% in ventricles from aging GDX mice. M-mode echocardiography showed no change in systolic function (e.g., ejection fraction). Critically, pulse-wave Doppler echocardiography showed that GDX slowed isovolumic relaxation time (12.9 ± 0.9 vs. 16.9 ± 1.0 ms, P < 0.05), indicative of diastolic dysfunction. Thus, dysregulation of intracellular Ca2+ and myofilament dysfunction contribute to deficits in contraction in hearts from testosterone-deficient aging mice. This suggests that low testosterone helps promote diastolic dysfunction in the aging heart. NEW & NOTEWORTHY The influence of long-term gonadectomy on contractile function was examined in aging male hearts. Gonadectomy slowed the decay of Ca2+ transients and contractions in ventricular myocytes and slowed isovolumic relaxation time, demonstrating diastolic dysfunction. Underlying mechanisms included Ca2+ dysregulation, elevated phospholamban protein levels, and hypophosphorylation of a myofilament protein, essential myosin light chain. Testosterone deficiency led to intracellular Ca2+ dysregulation and myofilament dysfunction, which may facilitate diastolic dysfunction in the setting of aging.
Collapse
Affiliation(s)
- Omar Ayaz
- Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia , Canada
| | - Shubham Banga
- Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia , Canada
| | - Stefan Heinze-Milne
- Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia , Canada
| | - Robert A Rose
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary , Calgary, Alberta , Canada
| | - W Glen Pyle
- Department of Biomedical Sciences, University of Guelph , Guelph, Ontario , Canada
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia , Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
41
|
Chaudhari S, Cushen SC, Osikoya O, Jaini PA, Posey R, Mathis KW, Goulopoulou S. Mechanisms of Sex Disparities in Cardiovascular Function and Remodeling. Compr Physiol 2018; 9:375-411. [PMID: 30549017 DOI: 10.1002/cphy.c180003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies demonstrate disparities between men and women in cardiovascular disease prevalence, clinical symptoms, treatments, and outcomes. Enrollment of women in clinical trials is lower than men, and experimental studies investigating molecular mechanisms and efficacy of certain therapeutics in cardiovascular disease have been primarily conducted in male animals. These practices bias data interpretation and limit the implication of research findings in female clinical populations. This review will focus on the biological origins of sex differences in cardiovascular physiology, health, and disease, with an emphasis on the sex hormones, estrogen and testosterone. First, we will briefly discuss epidemiological evidence of sex disparities in cardiovascular disease prevalence and clinical manifestation. Second, we will describe studies suggesting sexual dimorphism in normal cardiovascular function from fetal life to older age. Third, we will summarize and critically discuss the current literature regarding the molecular mechanisms underlying the effects of estrogens and androgens on cardiac and vascular physiology and the contribution of these hormones to sex differences in cardiovascular disease. Fourth, we will present cardiovascular disease risk factors that are positively associated with the female sex, and thus, contributing to increased cardiovascular risk in women. We conclude that inclusion of both men and women in the investigation of the role of estrogens and androgens in cardiovascular physiology will advance our understanding of the mechanisms underlying sex differences in cardiovascular disease. In addition, investigating the role of sex-specific factors in the development of cardiovascular disease will reduce sex and gender disparities in the treatment and diagnosis of cardiovascular disease. © 2019 American Physiological Society. Compr Physiol 9:375-411, 2019.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Paresh A Jaini
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rachel Posey
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Keisa W Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
42
|
Bernasochi GB, Boon WC, Delbridge LMD, Bell JR. The myocardium and sex steroid hormone influences. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
43
|
Abstract
Blood, serum and plasma represent accessible sources of data about physiological and pathologic status. In arrhythmogenic cardiomyopathy (ACM), circulating nucleated cells are routinely used for detection of germinal genetic mutations. In addition, different biomarkers have been proposed for diagnostic purposes and for monitoring disease progression, including inflammatory cytokines, markers of myocardial dysfunction and damage, and microRNAs. This review summarizes the current information that can be retrieved from the blood of ACM patients and considers the future prospects. Improvements in current knowledge of circulating factors may provide noninvasive means to simplify and improve the diagnosis, prognosis prediction, and management of ACM patients.
Collapse
|
44
|
Baka T, Hodosy J, Krajcirovicova K, Repova K, Aziriova S, Domonkos E, Borbelyova V, Slavkovsky P, Zorad S, Celec P, Paulis L, Simko F. 17β-Estradiol treatment reversed left ventricular dysfunction in castrated male rats: an echocardiographic study. Can J Physiol Pharmacol 2018; 96:850-854. [DOI: 10.1139/cjpp-2017-0596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
No data are available on heart function in chronic testosterone deficiency and on the effect of estrogen treatment. Eighteen 4-week-old male Lewis rats were randomly divided into 3 groups (n = 6): 1 group of sham-operated rats and 2 groups of castrated rats. Sixty-six weeks after surgery, 1 castrated group received a dose of 17β-estradiol (10 μg/kg per day) and the remaining 2 groups received a placebo subcutaneously for 14 days. Left ventricular (LV) systolic and diastolic functions were measured by transthoracic echocardiography. Castration decreased LV ejection fraction (9%) and fractional shortening (15%) and deteriorated LV diastolic function (94%). 17β-Estradiol treatment increased LV ejection fraction (15%) and fractional shortening (31%) and improved LV diastolic function (48%). Plasma testosterone concentrations were decreased in both castrated groups. In conclusion, chronic testosterone deficiency induced LV systolic and diastolic dysfunction; these disorders were reversed by short-term treatment with 17β-estradiol.
Collapse
Affiliation(s)
- Tomas Baka
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Julius Hodosy
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Kristina Krajcirovicova
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Kristina Repova
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Silvia Aziriova
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Emese Domonkos
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Veronika Borbelyova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Peter Slavkovsky
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Stefan Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovak Republic
| | - Peter Celec
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Ludovit Paulis
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71 Bratislava, Slovak Republic
| | - Fedor Simko
- Department of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovak Republic
- 3rd Clinic of Medicine, Faculty of Medicine, Comenius University, Limbova 5, 833 05 Bratislava, Slovak Republic
| |
Collapse
|
45
|
Jardí F, Laurent MR, Dubois V, Kim N, Khalil R, Decallonne B, Vanderschueren D, Claessens F. Androgen and estrogen actions on male physical activity: a story beyond muscle. J Endocrinol 2018; 238:R31-R52. [PMID: 29743340 DOI: 10.1530/joe-18-0125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022]
Abstract
Physical inactivity is a pandemic that contributes to several chronic diseases and poses a significant burden on health care systems worldwide. The search for effective strategies to combat sedentary behavior has led to an intensification of the research efforts to unravel the biological substrate controlling activity. A wide body of preclinical evidence makes a strong case for sex steroids regulating physical activity in both genders, albeit the mechanisms implicated remain unclear. The beneficial effects of androgens on muscle as well as on other peripheral functions might play a role in favoring adaptation to exercise. Alternatively or in addition, sex steroids could act on specific brain circuitries to boost physical activity. This review critically discusses the evidence supporting a role for androgens and estrogens stimulating male physical activity, with special emphasis on the possible role of peripheral and/or central mechanisms. Finally, the potential translation of these findings to humans is briefly discussed.
Collapse
Affiliation(s)
- Ferran Jardí
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Michaël R Laurent
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Gerontology and GeriatricsDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Vanessa Dubois
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nari Kim
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
46
|
Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 2018; 9:400-421. [PMID: 29469650 PMCID: PMC6219642 DOI: 10.1080/19490976.2018.1441664] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/24/2017] [Accepted: 02/08/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Conditions of excess androgen in women, such as polycystic ovary syndrome (PCOS), often exhibit intergenerational transmission. One way in which the risk for PCOS may be increased in daughters of affected women is through exposure to elevated androgens in utero. Hyperandrogenemic conditions have serious health consequences, including increased risk for hypertension and cardiovascular disease. Recently, gut dysbiosis has been found to induce hypertension in rats, such that blood pressure can be normalized through fecal microbial transplant. Therefore, we hypothesized that the hypertension seen in PCOS has early origins in gut dysbiosis caused by in utero exposure to excess androgen. We investigated this hypothesis with a model of prenatal androgen (PNA) exposure and maternal hyperandrogenemia by single-injection of testosterone cypionate or sesame oil vehicle (VEH) to pregnant dams in late gestation. We then completed a gut microbiota and cardiometabolic profile of the adult female offspring. RESULTS The metabolic assessment revealed that adult PNA rats had increased body weight and increased mRNA expression of adipokines: adipocyte binding protein 2, adiponectin, and leptin in inguinal white adipose tissue. Radiotelemetry analysis revealed hypertension with decreased heart rate in PNA animals. The fecal microbiota profile of PNA animals contained higher relative abundance of bacteria associated with steroid hormone synthesis, Nocardiaceae and Clostridiaceae, and lower abundance of Akkermansia, Bacteroides, Lactobacillus, Clostridium. The PNA animals also had an increased relative abundance of bacteria associated with biosynthesis and elongation of unsaturated short chain fatty acids (SCFAs). CONCLUSIONS We found that prenatal exposure to excess androgen negatively impacted cardiovascular function by increasing systolic and diastolic blood pressure and decreasing heart rate. Prenatal androgen was also associated with gut microbial dysbiosis and altered abundance of bacteria involved in metabolite production of short chain fatty acids. These results suggest that early-life exposure to hyperandrogenemia in daughters of women with PCOS may lead to long-term alterations in gut microbiota and cardiometabolic function.
Collapse
Affiliation(s)
- Shermel B. Sherman
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nadeen Sarsour
- Department of Biological Sciences, University of Toledo, Toledo, OH
| | - Marziyeh Salehi
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Allen Schroering
- Department of Neurosciences and Neurological Disorders, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Blair Mell
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Bina Joe
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Diabetes and Endocrine Research, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| |
Collapse
|
47
|
Chomphoo S, Pakkarato S, Sawatpanich T, Sakagami H, Kondo H, Hipkaeo W. Localization of EFA6 (exchange factor for ARF6) isoform D in steroidogenic testicular Leydig cells of adult mice. Acta Histochem 2018; 120:263-268. [PMID: 29496264 DOI: 10.1016/j.acthis.2018.02.009] [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: 01/07/2018] [Revised: 02/03/2018] [Accepted: 02/21/2018] [Indexed: 11/19/2022]
Abstract
EFA6 (exchange factor for ARF6) activates Arf6 (ADP ribosylation factor 6) by exchanging ADP to ATP and the resulting activated form of Arf6 is involved in the membrane trafficking and actin remodeling of cells. Our previous study has shown the selective expression/localization of EFA6D in steroidogenic adrenocortical cells in situ of adult mice. In view of the previous finding, the present study was undertaken to examine its localization in mouse Leydig cells representing another steroidogenic cell species in order to further support the possible involvement of the EFA6/Arf6 cascade via membrane trafficking in the regulation of steroidogenesis and/or secretion. A distinct band for EFA6D with the same size as that of the brain was detected in the testis of adult mice. In immuno-light microscopy, immunoreactivity for EFA6D was seen throughout the cytoplasm in most Leydig cells without any distinct accumulation along the plasmalemma. Lack of immunoreactivity for EFA6D was seen in the seminiferous tubular epithelium. In immuno-electron microscopy, the immune-labeling was seen in sporadic/focal patterns on plasma membranes and some vesicles and vacuoles subjacent to the plasma membranes. More constant and rather predominant is the labeling on numerous mitochondria. No immuno-labeling was seen in lipid droplets. The present study suggests that EFA6D is somehow involved in regulation of the synthesis and/or secretion of testosterone through the membrane-traffic by activation of Arf6. In addition, EFA6D is suggested to play in mitochondria some yet unidentified roles rather independent of Arf6-activation, which remains to be elucidated.
Collapse
Affiliation(s)
- Surang Chomphoo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sawetree Pakkarato
- Department of Social Sciences, Faculty of Sciences and Liberal Arts, Rajamangala University of Technology Isan, Sura Narai Rd, Nai-muang, Muang, Nakhon Ratchasima 30000, Thailand
| | - Tarinee Sawatpanich
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Hiroyuki Sakagami
- Department of Anatomy, School of Medicine, Kitasato University, Tokyo, Japan
| | - Hisatake Kondo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wiphawi Hipkaeo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
48
|
Akdis D, Saguner AM, Shah K, Wei C, Medeiros-Domingo A, von Eckardstein A, Lüscher TF, Brunckhorst C, Chen HSV, Duru F. Sex hormones affect outcome in arrhythmogenic right ventricular cardiomyopathy/dysplasia: from a stem cell derived cardiomyocyte-based model to clinical biomarkers of disease outcome. Eur Heart J 2018; 38:1498-1508. [PMID: 28329361 DOI: 10.1093/eurheartj/ehx011] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/10/2017] [Indexed: 12/11/2022] Open
Abstract
Aims Arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D) is characterized by fibrofatty infiltration of the myocardium and ventricular arrhythmias that may lead to sudden cardiac death. It has been observed that male patients develop the disease earlier and present with more severe phenotypes as compared to females. Thus, we hypothesized that serum levels of sex hormones may contribute to major arrhythmic cardiovascular events (MACE) in patients with ARVC/D. Methods and results The serum levels of five sex hormones, sex hormone-binding globulin, high sensitivity troponin T, pro-brain natriuretic peptide, cholesterol, triglycerides, insulin, and glucose were measured in 54 ARVC/D patients (72% male). Twenty-six patients (48%) experienced MACE. Total and free testosterone levels were significantly increased in males with MACE as compared to males with a favourable outcome, whereas estradiol was significantly lower in females with MACE as compared to females with a favourable outcome. Increased testosterone levels remained independently associated with MACE in males after adjusting for age, body mass index, Task Force criteria, ventricular function, and desmosomal mutation status. Furthermore, an induced pluripotent stem cell-derived ARVC/D cardiomyocyte model was used to investigate the effects of sex hormones. In this model, testosterone worsened and estradiol improved ARVC/D-related pathologies such as cardiomyocyte apoptosis and lipogenesis, strongly supporting our clinical findings. Conclusions Elevated serum testosterone levels in males and decreased estradiol levels in females are independently associated with MACE in ARVC/D, and directly influence disease pathology. Therefore, determining the levels of sex hormones may be useful for risk stratification and may open a new window for preventive interventions.
Collapse
Affiliation(s)
- Deniz Akdis
- Department of Cardiology, University Heart Center Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Ardan M Saguner
- Department of Cardiology, University Heart Center Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Khooshbu Shah
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Chuanyu Wei
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | - Arnold von Eckardstein
- Department of Clinical Chemistry, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Winterthurerstr 190, 8057 Zurich, Switzerland
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Winterthurerstr 190, 8057 Zurich, Switzerland
| | - Corinna Brunckhorst
- Department of Cardiology, University Heart Center Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - H S Vincent Chen
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Firat Duru
- Department of Cardiology, University Heart Center Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Winterthurerstr 190, 8057 Zurich, Switzerland
| |
Collapse
|
49
|
Ribeiro Júnior RF, Ronconi KS, Jesus ICG, Almeida PWM, Forechi L, Vassallo DV, Guatimosim S, Stefanon I, Fernandes AA. Testosterone deficiency prevents left ventricular contractility dysfunction after myocardial infarction. Mol Cell Endocrinol 2018; 460:14-23. [PMID: 28606867 DOI: 10.1016/j.mce.2017.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 05/17/2017] [Accepted: 06/08/2017] [Indexed: 11/30/2022]
Abstract
Testosterone may affect myocardial contractility since its deficiency decreases the contraction and relaxation of the heart. Meanwhile, testosterone replacement therapy has raised concerns because it may worsen cardiac dysfunction and remodeling after myocardial infarction (MI). In this study, we evaluate cardiac contractility 60 days after MI in rats with suppressed testosterone. Male Wistar rats underwent bilateral orchidectomy one week before the ligation of the anterior descending left coronary artery. The animals were divided into orchidectomized (OCT); MI; orchidectomized + MI (OCT + MI); orchidectomized + MI + testosterone (OCT + MI + T) and control (Sham) groups. Eight weeks after MI, papillary muscle contractility was analyzed under increasing calcium (0.62, 1.25, 2.5 and 3.75 mM) and isoproterenol (10-8 to 10-2 M) concentrations. Ventricular myocytes were isolated for intracellular calcium measurements and assessment of Ca2+ handling proteins. Contractility was preserved in the orchidectomized animals after myocardial infarction and was reduced when testosterone was replaced (Ca2+ 3.75 mM: Sham: 608 ± 70 (n = 11); OCT: 590 ± 37 (n = 16); MI: 311 ± 33* (n = 9); OCT + MI: 594 ± 76 (n = 7); OCT + MI + T: 433 ± 38* (n=4), g/g *p < 0.05 vs Sham). Orchidectomy also increased the Ca2+ transient amplitude of the ventricular myocytes and SERCA-2a protein expression levels. PLB phosphorylation levels at Thr17 were not different in the orchidectomized animals compared to the Sham animals but were reduced after testosterone replacement. CAMKII phosphorylation and protein nitrosylation increased in the orchidectomized animals. Our results support the view that testosterone deficiency prevents MI contractility dysfunction by altering the key proteins involved in Ca2+ handling.
Collapse
Affiliation(s)
- R F Ribeiro Júnior
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | - K S Ronconi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - I C G Jesus
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - P W M Almeida
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - L Forechi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - D V Vassallo
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - S Guatimosim
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - I Stefanon
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - A A Fernandes
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
50
|
Kerkhof PLM, Kuznetsova T, Yasha Kresh J, Handly N. Cardiophysiology Illustrated by Comparing Ventricular Volumes in Healthy Adult Males and Females. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:123-138. [PMID: 30051381 DOI: 10.1007/978-3-319-77932-4_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Recent advances in cardiac imaging techniques have substantially contributed to a growing interest in the analysis of global cardiac chamber dimensions and regional myocardial deformation. During the cardiac cycle, ventricular luminal volume varies due to the contraction process, which also confers a shape change including substantial alteration of long axis length, as well as rotation of the base compared to the apex. Local deformation can be assessed by strain (rate) analysis. Reviewing the present literature, it must be concluded that there is no single metric available to comprehensively characterize ventricular function. Every candidate advanced thus far has been found to incompletely reflect ventricular performance. This observation is not surprising in view of the complexity of the cardiac pump system. Additionally, sex-specific modifiers may play a role. More than three decades ago, it was shown that on average the ventricular volume is smaller in healthy women compared to matched males. Therefore, the present contribution concerns the interpretation of data derived from the healthy heart in both men and women. Starting from the classical Starling concept, we apply a simple mathematical transformation which permits an insightful representation of ventricular mechanics. Relating end-systolic volume (ESV) to end-diastolic volume creates the ventricular volume regulation graph which features the pertinent working point of an individual heart. This fundamental approach illustrates why certain proposed performance indexes cannot individually reveal the essence of ventricular systolic function. We demonstrate that particular metrics are highly interconnected and just tell us the same story in a different disguise. It is imperative to understand which associations exist and if they expectedly are (nearly) linear or frankly nonlinear. Notably, ejection fraction (EF) is primarily determined by ESV, while in turn EF is not much different from ventriculo-arterial coupling (VAC). Insight into cardiac function is promoted by identification of the paramount/essential components involved. The smaller ESV (p < 0.0001) implies that EF is higher in women and may also have consequences for VAC.
Collapse
Affiliation(s)
- Peter L M Kerkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands.
| | - Tatiana Kuznetsova
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - J Yasha Kresh
- Departments of Cardiothoracic Surgery and Medicine (Cardiology), Drexel University College of Medicine, IME, University of Pennsylvania, Philadelphia, PA, USA
| | - Neal Handly
- Department of Emergency Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|