1
|
Faakye A, Harold KM, Matsuzaki S, Pranay A, Mendez Garcia MF, Loveland BL, Rigsby SN, Peelor FF, Eyster C, Miller BF, Griffin TM, Kinter M, Chiao YA, Humphries KM. The effect of enhanced glycolysis on cardiac aging. GeroScience 2025:10.1007/s11357-025-01656-z. [PMID: 40310487 DOI: 10.1007/s11357-025-01656-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Cardiac aging is associated with metabolic changes, including an increased reliance on glycolysis, and an increased susceptibility to cardiovascular diseases. This study explores the relationship between enhanced cardiac glycolysis and aging using the GlycoHi mouse model, characterized by constitutively elevated glycolysis. We compared cardiac function, metabolism, mitochondrial performance, and hallmarks of aging between aged (21 and 24 months) GlycoHi and wild-type (WT) mice across sexes. Our findings reveal modest reductions in cardiac function in aged GlycoHi mice compared to WT mice, with sex-specific differences in heart size and collagen concentration. Female GlycoHi hearts exhibited hypertrophy without fibrosis, while males showed elevated collagen levels. Whole-body metabolic assessments revealed similar energy expenditure and respiratory patterns across genotypes, with females displaying less circadian-associated variation in metabolism. Mitochondrial analyses showed that aged GlycoHi hearts maintained metabolic adaptations favoring glycolysis but did not exhibit significant bioenergetic dysfunction or oxidative stress. Pyruvate dehydrogenase activity, initially elevated in younger GlycoHi hearts, normalized to WT levels with age. Proteomic and metabolomic analyses highlighted distinct profiles between genotypes, with GlycoHi hearts exhibiting increased glycolytic enzyme levels and reduced abundance of fatty acid oxidation proteins. Despite these differences, indicators of oxidative stress, proteostasis, and cellular senescence were comparable between genotypes, suggesting no acceleration of aging-related dysfunction. This study demonstrates that increased cardiac glycolysis alone does not suffice to drive accelerated cardiac aging. Instead, metabolic and functional changes in aged GlycoHi hearts reflect adaptations rather than pathological declines, providing insights into potential metabolic targets for interventions against cardiac aging.
Collapse
Affiliation(s)
- Anna Faakye
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kylene M Harold
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Maria F Mendez Garcia
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Brooke L Loveland
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Sandra N Rigsby
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Craig Eyster
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma City Veterans Association Medical Center, Oklahoma City, OK, USA
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13 th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
2
|
Holody CD, Woodman AG, Nie C, Liu SN, Young D, Wiedemeyer A, Carpenter R, Noble RMN, Graf D, Dufour A, Lemieux H, Bourque SL. Perinatal iron deficiency alters the cardiac proteome and mitochondrial function in neonatal offspring. Am J Physiol Heart Circ Physiol 2025; 328:H101-H112. [PMID: 39570196 DOI: 10.1152/ajpheart.00412.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
Iron deficiency (ID) is common during gestation and early infancy and can alter developmental trajectories with lasting consequences on cardiovascular health. Iron plays a critical role in systemic oxygen transport (via hemoglobin) and aerobic respiration (as a component of mitochondrial complexes). Perinatal ID has been shown to cause cardiac dysfunction in neonates, but the mechanisms underlying these changes have not been characterized. Here, we examined the effects of perinatal ID on cardiac mitochondrial function in rats in the early postnatal period. Female rats were fed an iron-restricted or iron-replete diet before and during pregnancy. Offspring hearts were collected postmortem for quantitative shotgun proteomic analysis [postnatal days (PD) 0 and 28] and mitochondrial function was assessed by high-resolution respirometry (at PD 0, 14, and 28). Markers of oxidative stress were measured by fluorescence microscopy and assessment of antioxidant gene expression profiles. Both male and female ID pups had reduced body weight and increased relative heart weights at all time points assessed, despite recovering from anemia by PD28. Proteomics analysis revealed dysregulation of mitochondrial proteins by ID, and these differences were most pronounced in males. In male hearts, ID increased mitochondrial content and decreased normalized mitochondrial respiration through the NADH-pathway, succinate-pathway, and fatty acid oxidation (FAO)-pathway. In conclusion, ID causes changes in cardiac mitochondrial function in neonates, which may reflect inadequate or maladaptive compensation during the transition from intrauterine to extrauterine life. Furthermore, the results presented herein, which were stratified by offspring sex, underscore the need for follow-up studies to directly assess differences in how male and female offspring cope with ID as a perinatal stressor.NEW & NOTEWORTHY Iron deficiency (ID) is the most common nutritional deficiency worldwide and is highly prevalent among pregnant women and young children. ID causes changes in mitochondrial protein expression and function in neonatal hearts, which may contribute to functional impairments. Improving cardiac energy metabolism may represent a novel approach to improve short- and long-term outcomes in infants affected by ID, but sex of the neonate may be an important determinant of treatment efficacy.
Collapse
Affiliation(s)
- Claudia D Holody
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew G Woodman
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Chunpeng Nie
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Si Ning Liu
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Young
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Alyssa Wiedemeyer
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rowan Carpenter
- Faculty Saint-Jean, University of Alberta, Edmonton, Alberta, Canada
| | - Ronan M N Noble
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Graf
- School of Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Hélène Lemieux
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty Saint-Jean, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Stephane L Bourque
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Kim SR, Kim MN, Cho DH, Kim HD, Bae SA, Kim HL, Kim MA, Hong KS, Shim WJ, Park SM. Sex differences of sequential changes in coronary blood flow and microvascular function in patients with suspected angina. Clin Res Cardiol 2024; 113:1638-1649. [PMID: 38112743 PMCID: PMC11579202 DOI: 10.1007/s00392-023-02358-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
AIMS This study evaluated the sex differences of sequential changes in coronary blood flows and microvascular function in patients with suspected angina but with no obstructed coronary arteries. METHODS A total of 202 consecutive patients who experienced chest pain but had no significant coronary artery stenosis and who underwent adenosine stress echocardiography were included in the study. Coronary blood flow (CBF) velocities were measured at 1, 2, and 3 min after adenosine infusion. RESULTS The mean age was 61 years, and 138 (68%) were women. Approximately 40% of patients had coronary microvascular dysfunction (CMD, coronary flow velocity reserve < 2.3), with women exhibiting higher CMD prevalence. The left ventricular (LV) mass index was similar between men and women, while women exhibited higher baseline rate pressure products (RPP). At baseline, coronary blood flow velocities were similar between the sexes. However, CBF velocities in women gradually increased during the examination; and in men, the increase was abrupt and steep during the early stages of examination (p = 0.015 for interaction between time and sex), even with similar RPP in stress. Coronary flow velocity reserve was steadily lower in women compared to men (1 min, 2.09 ± 0.86 vs 2.44 ± 0.87; 2 min, 2.39 ± 0.72 vs 2.63 ± 0.85; 3 min, 2.45 ± 0.70 vs 2.68 ± 0.73). CONCLUSIONS In patients with suspected angina but with no obstructed coronary arteries, CMD was especially prevalent among women. Women exhibited higher oxygen consumption, while exhibiting slower and gradual increases in CBF velocities. Conversely, men exhibited faster and steeper increases in CBF velocities even with similar RPP in stress.
Collapse
Affiliation(s)
- So Ree Kim
- Division of Cardiology, Korea University Anam Hospital, 73 Goryeodae-ro Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Mi-Na Kim
- Division of Cardiology, Korea University Anam Hospital, 73 Goryeodae-ro Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Dong-Hyuk Cho
- Division of Cardiology, Korea University Anam Hospital, 73 Goryeodae-ro Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hee-Dong Kim
- Division of Cardiology, Soonchunhyang University Hospital, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Sung A Bae
- Division of Cardiology, Yongin Severance Hospital, 363, Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Hack-Lyoung Kim
- Cardiovascular Center, Seoul National University Boramae Hospital, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, Republic of Korea
| | - Myung-A Kim
- Cardiovascular Center, Seoul National University Boramae Hospital, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, Republic of Korea
| | - Kyung-Soon Hong
- Division of Cardiology, Hallym University Chuncheon Sacred Heart Hospital, 77, Sakju-ro, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Wan Joo Shim
- Division of Cardiology, Korea University Anam Hospital, 73 Goryeodae-ro Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seong-Mi Park
- Division of Cardiology, Korea University Anam Hospital, 73 Goryeodae-ro Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
4
|
Shasteen ME, Wurzelmann MK, McGregor AJ, Raukar NP. Heart Breaking Differences: A Narrative Review of Sex and Gender Disparities in Sports-Related Sudden Cardiac Death. Clin Ther 2024; 46:982-987. [PMID: 39613695 DOI: 10.1016/j.clinthera.2024.11.002] [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: 04/08/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 12/01/2024]
Abstract
PURPOSE Sports-related sudden cardiac death (srSCD) represents a rare yet significant occurrence. This review aims to explore the epidemiology, etiology, and prevention of srSCD, with a particular focus on the influence of sex and gender. It seeks to analyze existing literature to elucidate the impact of biological variables, societal factors, and preventive measures in understanding and addressing srSCD among athletes. METHODS A narrative review approach was utilized to synthesize relevant literature on srSCD, using a validated PubMed Search tool for sex and gender-related factors. The review focused on primary data investigating sex differences that may contribute to srSCD, as well as pertinent review articles. FINDINGS The review highlights the complexity of defining and studying srSCD, with challenges stemming from varied reporting methods and lack of standardized definitions. Disparities in incidence rates between male and female athletes are evident, with males exhibiting a disproportionately higher risk. Biological factors, including cardiac adaptations to exercise and sex hormone influences, contribute to these sex-specific differences in srSCD rates. While screening programs, particularly utilizing electrocardiograms, show promise in identifying at-risk individuals, debates persist regarding their implementation and efficacy. Furthermore, legislative gaps in mandating the availability of automatic external defibrillators (AEDs) in public settings underscore the need for unified advocacy efforts to improve access to life-saving interventions. IMPLICATIONS Understanding the multifaceted nature of srSCD, including its biological underpinnings and societal implications, is crucial for developing effective preventive strategies. Sex-specific screening programs tailored to the unique risk profiles of male and female athletes, as well as legislative initiatives promoting AED placement and cardiopulmonary resuscitation training, are essential components of comprehensive srSCD prevention efforts. By addressing disparities and implementing evidence-based interventions, this paper advocates for a holistic approach to mitigate the risk of srSCD and enhance the safety and well-being of athletes across all levels of competition.
Collapse
Affiliation(s)
- Mallory E Shasteen
- Department of Emergency Medicine, University of South Carolina School of Medicine Greenville, Prisma Health-Upstate.
| | - Mary K Wurzelmann
- Department of Emergency Medicine, University of South Carolina School of Medicine Greenville, Prisma Health-Upstate
| | - Alyson J McGregor
- Department of Emergency Medicine, University of South Carolina School of Medicine Greenville, Prisma Health-Upstate
| | - Neha P Raukar
- Department of Emergency Medicine, Mayo Clinic College of Medicine
| |
Collapse
|
5
|
Jovanovic N, Zach V, Crocini C, Bahr LS, Forslund-Startceva SK, Franz K. A gender perspective on diet, microbiome, and sex hormone interplay in cardiovascular disease. Acta Physiol (Oxf) 2024; 240:e14228. [PMID: 39263901 DOI: 10.1111/apha.14228] [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: 03/29/2024] [Revised: 07/26/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024]
Abstract
A unique interplay between body and environment embeds and reflects host-microbiome interactions that contribute to sex-differential disease susceptibility, symptomatology, and treatment outcomes. These differences derive from individual biological factors, such as sex hormone action, sex-divergent immune processes, X-linked gene dosage effects, and epigenetics, as well as from their interaction across the lifespan. The gut microbiome is increasingly recognized as a moderator of several body systems that are thus impacted by its function and composition. In humans, biological sex components further interact with gender-specific exposures such as dietary preferences, stressors, and life experiences to form a complex whole, requiring innovative methodologies to disentangle. Here, we summarize current knowledge of the interactions among sex hormones, gut microbiota, immune system, and vascular health and their relevance for sex-differential epidemiology of cardiovascular diseases. We outline clinical implications, identify knowledge gaps, and place emphasis on required future studies to address these gaps. In addition, we provide an overview of the caveats associated with conducting cardiovascular research that require consideration of sex/gender differences. While previous work has inspected several of these components separately, here we call attention to further translational utility of a combined perspective from cardiovascular translational research, gender medicine, and microbiome systems biology.
Collapse
Affiliation(s)
- Nina Jovanovic
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Veronika Zach
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Claudia Crocini
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lina Samira Bahr
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sofia Kirke Forslund-Startceva
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Kristina Franz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| |
Collapse
|
6
|
Wang Q, Wang H, Du B, Wu Y, Li Z, Niu Y, Ouyang F, Bai K, Wang J, Sun K. Association between physical activity and cardiovascular parameters in 7-year-old children: a Chinese cross-sectional study. BMC Pediatr 2024; 24:522. [PMID: 39138455 PMCID: PMC11320774 DOI: 10.1186/s12887-023-04468-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 12/06/2023] [Indexed: 08/15/2024] Open
Abstract
INTRODUCTION Physical activity (PA) is believed to play an important part in many aspects during childhood and adolescence, especially cardiorespiratory fitness and cardiometabolic health. However, whether different levels of PA in daily life influence the structure or function of heart in school-aged children remains unknown. We aimed to investigate the association between PA and cardiovascular parameters in 7-year-old children. METHODS Follow-up data from the Shanghai Prenatal Cohort Study and the Shanghai Birth Cohort was analyzed. Perinatal information including both maternal and offspring datum was recorded. A refined questionnaire was used to evaluate the frequency and duration of children's PA levels. Blood pressure, echocardiography, and anthropometry assessment were conducted during the follow-up of 7-year-old children. RESULTS Overall, high PA level was associated with higher left ventricle posterior wall thickness in diastole (LVPWd, β coefficient: 0.36, 95% CI: 0.12, 0.61), higher left ventricle mass index (LVMI, β = 0.28, 95% CI: 0.07, 0.48), mitral E/a ratio (β = 0.47, 95% CI: 0.22, 0.71) and slower heart rate (β = -0.32, 95% CI: -0.57, -0.07), compared to low PA level. Medium PA level was associated with lower diastolic blood pressure (DBP, β = -0.18, 95% CI: -0.35, -0.01). In subgroup analysis, increased relative wall thickness (RWT) was found in high PA level boys (β = 0.36, 95% CI: 0.05, 0.67), and systolic blood pressure (SBP) showed a significant decrease in high PA level girls (β = -0.42, 95% CI: -0.78, -0.06). CONCLUSIONS This study suggested non-athlete children having higher PA level were associated with thicker left ventricle (LV) walls and better LV diastolic function, as well as slower heart rate and DBP at the age of 7. Furthermore, disparity in the association between PA level with morphological heart patterns and blood pressure existed in different sex category.
Collapse
Affiliation(s)
- Qianchuo Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Hualin Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Bowen Du
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yujian Wu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Zhuoyan Li
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yiwei Niu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Fengxiu Ouyang
- Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Bai
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| | - Jian Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
7
|
George SA, Trampel KA, Brunner K, Efimov IR. Moderate Endurance Exercise Increases Arrhythmia Susceptibility and Modulates Cardiac Structure and Function in a Sexually Dimorphic Manner. J Am Heart Assoc 2024; 13:e033317. [PMID: 38686869 PMCID: PMC11179941 DOI: 10.1161/jaha.123.033317] [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: 10/31/2023] [Accepted: 02/05/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Although moderate endurance exercise has been reported to improve cardiovascular health, its effects on cardiac structure and function are not fully characterized, especially with respect to sexual dimorphism. We aimed to assess the effects of moderate endurance exercise on cardiac physiology in male versus female mice. METHODS AND RESULTS C57BL/6J mice of both sexes were run on a treadmill for 6 weeks. ECG and echocardiography were performed every 2 weeks. After 6 weeks of exercise, mice were euthanized, and triple parametric optical mapping was performed on Langendorff perfused hearts to assess cardiac electrophysiology. Arrhythmia inducibility was tested by programmed electrical stimulation. Left ventricular tissue was fixed, and RNA sequencing was performed to determine exercise-induced transcriptional changes. Exercise-induced left ventricular dilatation was observed in female mice alone, as evidenced by increased left ventricular diameter and reduced left ventricular wall thickness. Increased cardiac output was also observed in female exercised mice but not males. Optical mapping revealed further sexual dimorphism in exercise-induced modulation of cardiac electrophysiology. In female mice, exercise prolonged action potential duration and reduced voltage-calcium influx delay. In male mice, exercise reduced the calcium decay constant, suggesting faster calcium reuptake. Exercise increased arrhythmia inducibility in both male and female mice; however, arrhythmia duration was increased only in females. Lastly, exercise-induced transcriptional changes were sex dependent: females and males exhibited the most significant changes in contractile versus metabolism-related genes, respectively. CONCLUSIONS Our data suggest that moderate endurance exercise can significantly alter multiple aspects of cardiac physiology in a sex-dependent manner. Although some of these effects are beneficial, like improved cardiac mechanical function, others are potentially proarrhythmic.
Collapse
Affiliation(s)
- Sharon A. George
- Department of Biomedical EngineeringGeorge Washington UniversityWashingtonDC
- Department of Biomedical EngineeringNorthwestern UniversityChicagoIL
| | - Katy Anne Trampel
- Department of Biomedical EngineeringGeorge Washington UniversityWashingtonDC
- Department of Biomedical EngineeringNorthwestern UniversityChicagoIL
| | - Kelsey Brunner
- Department of Biomedical EngineeringGeorge Washington UniversityWashingtonDC
| | - Igor R. Efimov
- Department of Biomedical EngineeringGeorge Washington UniversityWashingtonDC
- Department of Biomedical EngineeringNorthwestern UniversityChicagoIL
- Department of MedicineNorthwestern UniversityChicagoIL
| |
Collapse
|
8
|
Keuning ZA, Hendriks PM, Duijnhouwer AL, Meccanici F, Siebelink HMJ, van den Hoven AT, Geenen LW, Eindhoven JA, Baggen VJM, Cuypers JAAE, Kauling RM, Roos-Hesselink JW, van den Bosch AE. Blood and Imaging Biomarkers in the Long-term Follow-up of Bicuspid Aortic Valve Patients. CJC Open 2024; 6:1-10. [PMID: 38313345 PMCID: PMC10837668 DOI: 10.1016/j.cjco.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/20/2023] [Indexed: 02/06/2024] Open
Abstract
Background Bicuspid aortic valve (BAV) is a common congenital heart defect. Patients with BAV are at risk for long-term complications such as valve stenosis and regurgitation. This study aimed to investigate sex differences in blood and imaging biomarkers and to describe the long-term prognostic value of blood and echocardiographic biomarkers. Methods Patients were included from 2 prospective observational cohort studies; they underwent venous blood sampling and transthoracic echocardiography including speckle tracking. Analyzed blood biomarkers were red-cell distribution width (RDW), creatinine, C-reactive protein (CRP), troponin T, N-terminal pro B-type natriuretic peptide (NT-proBNP), and transforming growth factor-beta (TGF-β). Sex differences were analyzed at baseline. Associations between biomarkers and arrhythmia-free and intervention-free survival were determined by Cox regression, adjusted for age and sex. Results A total of 182 patients with BAV were included: median age 34; interquartile range [IQR]: 23-46 years; 55.5% male. CRP, NT-proBNP, and RDW were higher in women, whereas creatinine, troponin T and TGF-β were higher among men. After a median follow-up time of 6.9 (IQR: 6.5-9.9) years, arrhythmia-free and intervention-free survival was, 81.0% and 73.1%, respectively. NT-proBNP was associated with both arrhythmia-free and intervention-free survival (hazard ratio [HR], 1.94, P = 0.005 and HR, 2.06, P = 0.002, respectively). On echocardiography higher left atrial (LA) size, left ventricular end-diastolic diameter (LVEDD), left ventricular (LV) mass index and E/e' ratio were associated with lower arrhythmia-free survival, whereas higher LA size, LV mass index, aortic valve peak velocity, and aortic regurgitation were associated with lower intervention-free survival. Conclusions Differences were observed in blood biomarkers between men and women with BAV. Besides LV systolic parameters, diastolic LV function and NT-proBNP should have a more prominent role as prognostic markers in clinical care.
Collapse
Affiliation(s)
- Zoë A Keuning
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Paul M Hendriks
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | - Frederike Meccanici
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | - Allard T van den Hoven
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Laurie W Geenen
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Jannet A Eindhoven
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Vivan J M Baggen
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Judith A A E Cuypers
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Robert M Kauling
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Jolien W Roos-Hesselink
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- ERN-GUARD-Heart: European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Amsterdam, The Netherlands
| | - Annemien E van den Bosch
- Department of Cardiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- ERN-GUARD-Heart: European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Chen HY, Gordon JW, Dwork N, Chung BT, Riselli A, Sivalokanathan S, Bok RA, Slater JB, Vigneron DB, Abraham MR, Larson PE. Probing Human Heart TCA Cycle Metabolism and Response to Glucose Load using Hyperpolarized [2- 13C]Pyruvate MR Spectroscopy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.16.23297053. [PMID: 37905131 PMCID: PMC10615004 DOI: 10.1101/2023.10.16.23297053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Introduction The normal heart has remarkable metabolic flexibility that permits rapid switching between mitochondrial glucose oxidation and fatty acid (FA) oxidation to generate ATP. Loss of metabolic flexibility has been implicated in the genesis of contractile dysfunction seen in cardiomyopathy. Metabolic flexibility has been imaged in experimental models, using hyperpolarized (HP) [2-13C]pyruvate MRI, which enables interrogation of metabolites that reflect tricarboxylic acid (TCA) cycle flux in cardiac myocytes. This study aimed to develop methods, demonstrate feasibility for [2-13C]pyruvate MRI in the human heart for the first time, and assess cardiac metabolic flexibility. Methods Good Manufacturing Practice [2-13C]pyruvic acid was polarized in a 5T polarizer for 2.5-3 hours. Following dissolution, QC parameters of HP pyruvate met all safety and sterility criteria for pharmacy release, prior to administration to study subjects. Three healthy subjects each received two HP injections and MR scans, first under fasting conditions, followed by oral glucose load. A 5cm axial slab-selective spectroscopy approach was prescribed over the left ventricle and acquired at 3s intervals on a 3T clinical MRI scanner. Results The study protocol which included HP substrate injection, MR scanning and oral glucose load, was performed safely without adverse events. Key downstream metabolites of [2-13C]pyruvate metabolism in cardiac myocytes include the glycolytic derivative [2-13C]lactate, TCA-associated metabolite [5-13C]glutamate, and [1-13C]acetylcarnitine, catalyzed by carnitine acetyltransferase (CAT). After glucose load, 13C-labeling of lactate, glutamate, and acetylcarnitine from 13C-pyruvate increased by 39.3%, 29.5%, and 114%, respectively in the three subjects, that could result from increases in lactate dehydrogenase (LDH), pyruvate dehydrogenase (PDH), and CAT enzyme activity as well as TCA cycle flux (glucose oxidation). Conclusions HP [2-13C]pyruvate imaging is safe and permits non-invasive assessment of TCA cycle intermediates and the acetyl buffer, acetylcarnitine, which is not possible using HP [1-13C]pyruvate. Cardiac metabolite measurement in the fasting/fed states provides information on cardiac metabolic flexibility and the acetylcarnitine pool.
Collapse
Affiliation(s)
- Hsin-Yu Chen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Jeremy W. Gordon
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Nicholas Dwork
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Brian T. Chung
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Andrew Riselli
- School of Pharmacy, University of California, San Francisco, United States
| | | | - Robert A. Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - James B. Slater
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Daniel B. Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - M. Roselle Abraham
- Division of Cardiology, University of California, San Francisco, United States
| | - Peder E.Z. Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| |
Collapse
|
10
|
Fliegner D, Ellieva A, Angelov A, Petrov G, Regitz-Zagrosek V. Sex differences and estrogen effects in cardiac mitochondria in human aortic stenosis and in the mouse heart. Front Endocrinol (Lausanne) 2023; 14:1181044. [PMID: 37916152 PMCID: PMC10617023 DOI: 10.3389/fendo.2023.1181044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Sex differences in the adaptation to pressure overload have been described in humans, as well as animal models, and have been related to sex-specific expression of mitochondrial genes. We therefore tested whether sex differences in cardiac mitochondrial respiration exist in humans with aortic stenosis (AS). We also examined whether these potential differences may be at least partially due to sex hormones by testing if mitochondrial respiration is affected by estrogen (17ß-estradiol (E2)). Methods Consecutive patients undergoing transapical aortic valve implantation (TAVI) (women, n = 7; men, n = 10) were included. Cardiac biopsies were obtained during TAVI and used directly for mitochondrial function measurements. Male and female C57BL/6J mice (n = 8/group) underwent sham surgery or gonadectomy (GDX) at the age of 2 months. After 14 days, mice were treated once with intraperitoneally injected vehicle (placebo), 17ß-estradiol (E2), estrogen receptor alpha (ERα) agonist [propyl pyrazole triol (PPT)], or ER beta (ERβ) agonist (BAY-1214257). Thereafter, mitochondrial measurements were performed directly in cardiac skinned fibers from isolated left ventricles and musculus solei. Results Mitochondrial State-3 respiration was higher in female than that in male human heart biopsies (15.0 ± 2.30 vs. 10.3 ± 2.05 nmol/mL/min/mg, p< 0.05). In the mouse model, mitochondrial State-3 respiration decreased significantly after GDX in female (27.6 ± 1.55 vs. 21.4 ± 1.71 nmol/mL/min/mg; p< 0.05) and male hearts (30.7 ± 1,48 vs. 23.7 ± 2,23 nmol/mL/min/mg; p< 0.05). In ovariectomized female mice, E2 and ERβ-agonist treatment restored the State-3 respiration to intact placebo level, whereas ERα-agonist treatment did not modulate State-3 respiration. The treatment with E2, ERα-, or ERβ-agonist did not modulate the State-3 respiration in GDX male mice. Conclusion We identified sex differences in mitochondrial respiration in the diseased human heart. This is in alignment with known sex differences in the gene expression and proteome level at the functional level. E2 and ERβ affect cardiac mitochondrial function in the mouse model, suggesting that they may also contribute to the sex differences in the human heart. Their roles should be further investigated.
Collapse
Affiliation(s)
- Daniela Fliegner
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
- Medical Affairs Internal Medicine, Pfizer Pharma GmbH, Berlin, Germany
| | - Alexandra Ellieva
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Angelov
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
- Clinic for Cardiology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
11
|
Popova OV, Rusanov VB. Is space flight arrhythmogenic? Front Physiol 2023; 14:1162355. [PMID: 37250132 PMCID: PMC10213435 DOI: 10.3389/fphys.2023.1162355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
|
12
|
Fairweather D, Beetler DJ, Musigk N, Heidecker B, Lyle MA, Cooper LT, Bruno KA. Sex and gender differences in myocarditis and dilated cardiomyopathy: An update. Front Cardiovasc Med 2023; 10:1129348. [PMID: 36937911 PMCID: PMC10017519 DOI: 10.3389/fcvm.2023.1129348] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
In the past decade there has been a growing interest in understanding sex and gender differences in myocarditis and dilated cardiomyopathy (DCM), and the purpose of this review is to provide an update on this topic including epidemiology, pathogenesis and clinical presentation, diagnosis and management. Recently, many clinical studies have been conducted examining sex differences in myocarditis. Studies consistently report that myocarditis occurs more often in men than women with a sex ratio ranging from 1:2-4 female to male. Studies reveal that DCM also has a sex ratio of around 1:3 women to men and this is also true for familial/genetic forms of DCM. Animal models have demonstrated that DCM develops after myocarditis in susceptible mouse strains and evidence exists for this progress clinically as well. A consistent finding is that myocarditis occurs primarily in men under 50 years of age, but in women after age 50 or post-menopause. In contrast, DCM typically occurs after age 50, although the age that post-myocarditis DCM occurs has not been investigated. In a small study, more men with myocarditis presented with symptoms of chest pain while women presented with dyspnea. Men with myocarditis have been found to have higher levels of heart failure biomarkers soluble ST2, creatine kinase, myoglobin and T helper 17-associated cytokines while women develop a better regulatory immune response. Studies of the pathogenesis of disease have found that Toll-like receptor (TLR)2 and TLR4 signaling pathways play a central role in increasing inflammation during myocarditis and in promoting remodeling and fibrosis that leads to DCM, and all of these pathways are elevated in males. Management of myocarditis follows heart failure guidelines and there are currently no disease-specific therapies. Research on standard heart failure medications reveal important sex differences. Overall, many advances in our understanding of the effect of biologic sex on myocarditis and DCM have occurred over the past decade, but many gaps in our understanding remain. A better understanding of sex and gender effects are needed to develop disease-targeted and individualized medicine approaches in the future.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Nicolas Musigk
- Department of Cardiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Heidecker
- Department of Cardiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Melissa A. Lyle
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
13
|
Zhou K, Xiao J, Wang H, Ni B, Huang J, Long X. Estradiol regulates oxidative stress and angiogenesis of myocardial microvascular endothelial cells via the CDK1/CDK2 pathway. Heliyon 2023; 9:e14305. [PMID: 36942258 PMCID: PMC10023923 DOI: 10.1016/j.heliyon.2023.e14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Cardiovascular diseases remain the leading cause of death, morbidity, and disability. Recently, it has been reported that gonadal hormones such as estradiol can act on membrane receptors and activate intracellular signaling mechanisms, thereby altering cellular function. This study aims to explore the function and molecular mechanism of estradiol on cardiac microvascular endothelial cells (CMVECs). Estradiol had low toxicity to CMVECs. Hypoxia/reoxygenation (H/R) stimulation inhibited the proliferation and migration of CMVECs, while estradiol significantly promoted proliferation and migration. Estradiol inhibited il-1, IL6, and TNF-α secretion levels after H/R stimulation. Meanwhile, estradiol inhibits oxidative stress and promotes angiogenesis. Further, estradiol upregulated the gene and protein levels of cyclin-dependent kinases 1 (CDK1) and CDK2 after H/R stimulation. When knocking down CDK1 and CDK2 of CMVECs, estradiol did not affect the protein expression of Cyclin E1 and Cyclin D1. Meanwhile, the regulatory effect of estradiol on oxidative stress, angiogenesis, and inflammatory response was significantly weakened or even disappeared. In conclusion, estradiol mediates oxidative stress and angiogenesis of myocardial microvascular endothelial cells by regulating the CDK/cyclin signaling pathway.
Collapse
Affiliation(s)
- Ke Zhou
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Jun Xiao
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
- Corresponding author.
| | - Hao Wang
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Bing Ni
- Institute of Immunology of Army Medical University, Chongqing, 400014, China
| | - Jietao Huang
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Xueyuan Long
- Vasculocardiology Department, Chongqing University Central Hospital, Chongqing, 400014, China
| |
Collapse
|
14
|
Sung KT, Chandramouli C, Lo CI, Tsai JP, Lai YH, Hsiao CC, Tsai SY, Yun CH, Hung TC, Kuo JY, Lin JL, Hou CJY, Chen YJ, Su CH, Hung CL, Bulwer BE, Yeh HI, Lam CSP. Association of Female Menopause With Atrioventricular Mechanics and Outcomes. Front Cardiovasc Med 2022; 9:804336. [PMID: 35528841 PMCID: PMC9068967 DOI: 10.3389/fcvm.2022.804336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDespite known sex differences in cardiac structure and function, little is known about how menopause and estrogen associate with atrioventricular mechanics and outcomes.ObjectiveTo study how, sex differences, loss of estrogen in menopause and duration of menopause, relate to atrioventricular mechanics and outcomes.MethodsAmong 4051 asymptomatic adults (49.8 ± 10.8 years, 35%women), left ventricular (LV) and left atrial (LA) mechanics were assessed using speckle-tracking.ResultsPost-menopausal (vs. pre-menopausal) women had similar LV ejection fraction but reduced GLS, reduced PALS, increased LA stiffness, higher LV sphericity and LV torsion (all p < 0.001). Multivariable analysis showed menopause to be associated with greater LV sphericity (0.02, 95%CI 0.01, 0.03), higher indexed LV mass (LVMi), lower mitral e’, lower LV GLS (0.37, 95%CI 0.04–0.70), higher LV torsion, larger LA volume, worse PALS (∼2.4-fold) and greater LA stiffness (0.028, 95%CI 0.01–0.05). Increasing years of menopause was associated with further reduction in GLS, markedly worse LA mechanics despite greater LV sphericity and higher torsion. Lower estradiol levels correlated with more impaired LV diastolic function, impaired LV GLS, greater LA stiffness, and increased LV sphericity and LV torsion (all p < 0.05). Approximately 5.5% (37/669) of post-menopausal women incident HF over 2.9 years of follow-up. Greater LV sphericity [adjusted hazard ratio (aHR) 1.04, 95%CI 1.00–1.07], impaired GLS (aHR 0.87, 95%CI 0.78–0.97), reduced peak left atrial longitudinal strain (PALS, aHR 0.94, 95%CI 0.90–0.99) and higher LA stiffness (aHR 10.5, 95%CI 1.69–64.6) were independently associated with the primary outcome of HF hospitalizations in post-menopause. Both PALS < 23% (aHR:1.32, 95%CI 1.01–3.49) and GLS < 16% (aHR:5.80, 95%CI 1.79–18.8) remained prognostic for the incidence of HF in post-menopausal women in dichotomous analyses, even after adjusting for confounders. Results were consistent with composite outcomes of HF hospitalizations and 1-year all-cause mortality as well.ConclusionMenopause was associated with greater LV/LA remodeling and reduced LV longitudinal and LA function in women. The cardiac functional deficit with menopause and lower estradiol levels, along with their independent prognostic value post-menopause, may elucidate sex differences in heart failure further.
Collapse
Affiliation(s)
- Kuo-Tzu Sung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan
| | - Chanchal Chandramouli
- National Heart Research Institute, National Heart Centre Singapore, Singapore, Singapore
- Academic Clinical Programme, Duke-National University of Singapore, Singapore, Singapore
- *Correspondence: Chanchal Chandramouli,
| | - Chi-In Lo
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jui-Peng Tsai
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Yau-Huei Lai
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chih-Chung Hsiao
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Shin-Yi Tsai
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Biomedical Imaging and Radiological Science, National Yang Ming University, Taipei, Taiwan
- School of Public Health, Johns Hopkins University Bloomberg, Baltimore, MD, United States
| | - Chun-Ho Yun
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Ta-Chuan Hung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Jen-Yuan Kuo
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Jiun-Lu Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Charles Jia-Yin Hou
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Ying-Ju Chen
- Department of Telehealth, MacKay Memorial Hospital, Taipei, Taiwan
| | - Cheng-Huang Su
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chung-Lieh Hung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Chung-Lieh Hung,
| | - Bernard E. Bulwer
- Brigham and Women’s Hospital, Boston, MA, United States
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Hung-I Yeh
- Academic Clinical Programme, Duke-National University of Singapore, Singapore, Singapore
- Department of Medicine, University Medical Centre Groningen, Groningen, Netherlands
| | - Carolyn S. P. Lam
- National Heart Research Institute, National Heart Centre Singapore, Singapore, Singapore
- Academic Clinical Programme, Duke-National University of Singapore, Singapore, Singapore
- Department of Medicine, University Medical Centre Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
St. Pierre SR, Peirlinck M, Kuhl E. Sex Matters: A Comprehensive Comparison of Female and Male Hearts. Front Physiol 2022; 13:831179. [PMID: 35392369 PMCID: PMC8980481 DOI: 10.3389/fphys.2022.831179] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/02/2022] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease in women remains under-diagnosed and under-treated. Recent studies suggest that this is caused, at least in part, by the lack of sex-specific diagnostic criteria. While it is widely recognized that the female heart is smaller than the male heart, it has long been ignored that it also has a different microstructural architecture. This has severe implications on a multitude of cardiac parameters. Here, we systematically review and compare geometric, functional, and structural parameters of female and male hearts, both in the healthy population and in athletes. Our study finds that, compared to the male heart, the female heart has a larger ejection fraction and beats at a faster rate but generates a smaller cardiac output. It has a lower blood pressure but produces universally larger contractile strains. Critically, allometric scaling, e.g., by lean body mass, reduces but does not completely eliminate the sex differences between female and male hearts. Our results suggest that the sex differences in cardiac form and function are too complex to be ignored: the female heart is not just a small version of the male heart. When using similar diagnostic criteria for female and male hearts, cardiac disease in women is frequently overlooked by routine exams, and it is diagnosed later and with more severe symptoms than in men. Clearly, there is an urgent need to better understand the female heart and design sex-specific diagnostic criteria that will allow us to diagnose cardiac disease in women equally as early, robustly, and reliably as in men. Systematic Review Registration https://livingmatter.stanford.edu/.
Collapse
Affiliation(s)
- Sarah R. St. Pierre
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | - Mathias Peirlinck
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
- Department of Biomechanical Engineering, Delft University of Technology, Delft, Netherlands
- Department of Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Ellen Kuhl
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
16
|
Crescioli C. The Role of Estrogens and Vitamin D in Cardiomyocyte Protection: A Female Perspective. Biomolecules 2021; 11:1815. [PMID: 34944459 PMCID: PMC8699224 DOI: 10.3390/biom11121815] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
Women experience a dramatical raise in cardiovascular events after menopause. The decline in estrogens is pointed to as the major responsible trigger for the increased risk of cardiovascular disease (CVD). Indeed, the menopausal transition associates with heart macro-remodeling, which results from a fine-tuned cell micro-remodeling. The remodeling of cardiomyocytes is a biomolecular response to several physiologic and pathologic stimuli, allowing healthy adaptation in normal conditions or maladaptation in an unfavorable environment, ending in organ architecture disarray. Estrogens largely impinge on cardiomyocyte remodeling, but they cannot fully explain the sex-dimorphism of CVD risk. Albeit cell remodeling and adaptation are under multifactorial regulation, vitamin D emerges to exert significant protective effects, controlling some intracellular paths, often shared with estrogen signaling. In post-menopause, the unfavorable association of hypoestrogenism-D hypovitaminosis may converge towards maladaptive remodeling and contribute to increased CVD risk. The aim of this review is to overview the role of estrogens and vitamin D in female cardiac health, speculating on their potential synergistic effect in cardiomyocyte remodeling, an issue that is not yet fully explored. Further learning the crosstalk between these two steroids in the biomolecular orchestration of cardiac cell fate during adaptation may help the translational approach to future cardioprotective strategies for women health.
Collapse
Affiliation(s)
- Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", 00135 Rome, Italy
| |
Collapse
|
17
|
Sex-Specific Impacts of Exercise on Cardiovascular Remodeling. J Clin Med 2021; 10:jcm10173833. [PMID: 34501285 PMCID: PMC8432130 DOI: 10.3390/jcm10173833] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/21/2021] [Accepted: 08/21/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in men and women. Biological sex plays a major role in cardiovascular physiology and pathological cardiovascular remodeling. Traditionally, pathological remodeling of cardiovascular system refers to the molecular, cellular, and morphological changes that result from insults, such as myocardial infarction or hypertension. Regular exercise training is known to induce physiological cardiovascular remodeling and beneficial functional adaptation of the cardiovascular apparatus. However, impact of exercise-induced cardiovascular remodeling and functional adaptation varies between males and females. This review aims to compare and contrast sex-specific manifestations of exercise-induced cardiovascular remodeling and functional adaptation. Specifically, we review (1) sex disparities in cardiovascular function, (2) influence of biological sex on exercise-induced cardiovascular remodeling and functional adaptation, and (3) sex-specific impacts of various types, intensities, and durations of exercise training on cardiovascular apparatus. The review highlights both animal and human studies in order to give an all-encompassing view of the exercise-induced sex differences in cardiovascular system and addresses the gaps in knowledge in the field.
Collapse
|
18
|
Zhu D, Chen W, Pan Y, Li T, Cui M, Chen B. The correlation between maternal age, parity, cardiac diastolic function and occurrence rate of pre-eclampsia. Sci Rep 2021; 11:8842. [PMID: 33893350 PMCID: PMC8065037 DOI: 10.1038/s41598-021-87953-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/17/2021] [Indexed: 01/04/2023] Open
Abstract
To evaluate the effect of age and parity on maternal cardiac diastolic function in second trimester among pregnant women with normal left ventricular ejection fraction. To analyze the correlation between impaired diastolic function and pre-eclampsia. It had been suggested that maternal cardiac adaptations during pregnancy differed between nulliparous and primiparous women and also varied according to age. Impaired cardiac function may precede pre-eclampsia. Therefore, we examined the effects of parity and age on cardiac diastolic function during pregnancy and whether impaired diastolic function during the second trimester correlates with pre-eclampsia. Women with singleton pregnancies at 13 + 0 to 27 + 6 weeks’ gestation and left ventricular ejection fraction (LVEF) ≥ 50% were retrospectively identified. Diastolic function parameters were assessed using transthoracic echocardiography. Pre-eclampsia was identified from medical records. The effect of age and parity on maternal cardiac diastolic function as well as the correlation between impaired diastolic function and occurrence rate of pre-eclampsia were examined. 376 pregnant women were included (median age: 30 years; median gestational age: 14 weeks; 171 primiparous women). LVEF was 66%. Impaired cardiac diastolic function was seen in 7.8% of pregnant women < 35 years compared with 28.6% of those ≥ 35 years (p = 0.000). ROC curve showed women with maternal age over 32 began to have a higher rate of impaired cardiac diastolic function (AUC = 0.704, p = 0.000, sensitivity = 54.5%, specificity = 75.3%). There was no difference in diastolic function indices between maternal women grouped by parity. Higher maternal age was an independent risk factor of declining Em (p < 0.05). Em < 13 cm/s was significantly associated with pre-eclampsia occurrence (HR 8.56; 95% CI 3.40–21.57) after being adjusted for confounders. Maternal age is an independent risk factor for diastolic function decline. There is no difference in cardiac diastolic function between nulliparous women and primiparous women. Pre-eclampsia occurrence is significantly higher in patients with impaired diastolic function at mid-gestation. The application of risk grading using diastolic function at mid-gestation may improve the survival outcomes of pregnant women.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| | - Weiyu Chen
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Yuchen Pan
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Tingcui Li
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ming Cui
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Baoxia Chen
- Department of Cardiology, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, 100191, China
| |
Collapse
|
19
|
Bugyei-Twum A. Sex differences in long-term effects of exertional heat stroke on myocardial metabolism. J Physiol 2020; 598:3829-3830. [PMID: 32598028 DOI: 10.1113/jp279984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 11/08/2022] Open
|
20
|
Laitano O, Garcia CK, Mattingly AJ, Robinson GP, Murray KO, King MA, Ingram B, Ramamoorthy S, Leon LR, Clanton TL. Delayed metabolic dysfunction in myocardium following exertional heat stroke in mice. J Physiol 2020; 598:967-985. [PMID: 32026469 DOI: 10.1113/jp279310] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Exposure to exertional heat stroke (EHS) is associated with increased risk of long-term cardiovascular disorders in humans. We demonstrate that in female mice, severe EHS results in metabolic changes in the myocardium, emerging only after 9-14 days. This was not observed in males that were symptom-limited at much lower exercise levels and heat loads compared to females. At 14 days of recovery in females, there were marked elevations in myocardial free fatty acids, ceramides and diacylglycerols, consistent with development of underlying cardiac abnormalities. Glycolysis shifted towards the pentose phosphate and glycerol-3-phosphate dehydrogenase pathways. There was evidence for oxidative stress, tissue injury and microscopic interstitial inflammation. The tricarboxylic acid cycle and nucleic acid metabolism pathways were also negatively affected. We conclude that exposure to EHS in female mice has the capacity to cause delayed metabolic disorders in the heart that could influence long-term health. ABSTRACT Exposure to exertional heat stroke (EHS) is associated with a higher risk of long-term cardiovascular disease in humans. Whether this is a cause-and-effect relationship remains unknown. We studied the potential of EHS to contribute to the development of a 'silent' form of cardiovascular disease using a preclinical mouse model of EHS. Plasma and ventricular myocardial samples were collected over 14 days of recovery. Male and female C57bl/6J mice underwent forced wheel running for 1.5-3 h in a 37.5°C/40% relative humidity until symptom limitation, characterized by CNS dysfunction. They reached peak core temperatures of 42.2 ± 0.3°C. Females ran ∼40% longer, reaching ∼51% greater heat load. Myocardial and plasma samples (n = 8 per group) were obtained between 30 min and 14 days of recovery, analysed using metabolomics/lipidomics platforms and compared to exercise controls. The immediate recovery period revealed an acute energy substrate crisis from which both sexes recovered within 24 h. However, at 9-14 days, the myocardium of female mice developed marked elevations in free fatty acids, ceramides and diacylglycerols. Glycolytic and tricarboxylic acid cycle metabolites revealed bottlenecks in substrate flow, with build-up of intermediate metabolites consistent with oxidative stress and damage. Males exhibited only late stage reductions in acylcarnitines and elevations in acetylcarnitine. Histopathology at 14 days showed interstitial inflammation in the female hearts only. The results demonstrate that the myocardium of female mice is vulnerable to a slowly emerging metabolic disorder following EHS that may harbinger long-term cardiovascular complications. Lack of similar findings in males may reflect their lower heat exposure.
Collapse
Affiliation(s)
- Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Christian K Garcia
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Alex J Mattingly
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Gerard P Robinson
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Kevin O Murray
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Michelle A King
- US Army Research Institute for Environmental Medicine, Natick, MA, USA
| | | | | | - Lisa R Leon
- US Army Research Institute for Environmental Medicine, Natick, MA, USA
| | - Thomas L Clanton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
21
|
Atrioventricular mechanical coupling and major adverse cardiac events in female patients following acute ST elevation myocardial infarction. Int J Cardiol 2019; 299:31-36. [PMID: 31300172 DOI: 10.1016/j.ijcard.2019.06.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND Sex-specific outcome data following myocardial infarction (MI) are inconclusive with some evidence suggesting association of female sex and increased major adverse cardiac events (MACE). Since mechanistic principles remain elusive, we aimed to quantify the underlying phenotype using cardiovascular magnetic resonance (CMR) quantitative deformation imaging and tissue characterisation. METHODS In total, 795 ST-elevation MI patients underwent post-interventional CMR imaging. Feature-tracking (CMR-FT) was performed in a blinded core-laboratory. Left ventricular function was quantified using ejection fraction (LVEF) and global longitudinal/circumferential/radial strains (GLS/GCS/GRS). Left atrial function was assessed by reservoir (εs), conduit (εe) and booster-pump strains (εa). Tissue characterisation included infarct size, microvascular obstruction and area at risk. Primary endpoint was the occurrence of MACE within 1 year. RESULTS Female sex was associated with increased MACE (HR 1.96, 95% CI 1.13-3.42, p = 0.017) but not independently of baseline confounders (p = 0.526) with women being older, more often diabetic and hypertensive (p < 0.001) and of higher Killip-class (p = 0.010). Tissue characterisation was similar between sexes. Women showed impaired atrial (εs p = 0.011, εe p < 0.001) but increased systolic ventricular mechanics (GLS p = 0.001, LVEF p = 0.048). While atrial and ventricular function predicted MACE in men only LV GLS and GCS were associated with MACE in women irrespective of confounders (GLS p = 0.036, GCS p = 0.04). CONCLUSION In men ventricular systolic contractility is impaired and volume assessments precisely stratify elevated risks. In contrast, women experience reduced atrial but increased ventricular systolic strain. This may reflect ventricular diastolic failure with systolic compensation, which is independently associated with MACE adding incremental value to sex-specific prognosis evaluation.
Collapse
|
22
|
Egelund J, Nyberg M, Mandrup CM, Abdulla J, Stallknecht B, Bangsbo J, Hellsten Y, Larsson HBW. Cardiac perfusion and function after high-intensity exercise training in late premenopausal and recent postmenopausal women: an MRI study. J Appl Physiol (1985) 2019; 126:1272-1280. [PMID: 30870082 DOI: 10.1152/japplphysiol.01089.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We examined the influence of recent menopause and aerobic exercise training in women on myocardial perfusion, left ventricular (LV) dimension, and function. Two groups (n = 14 each) of healthy late premenopausal (50.2 ± 2.1 yr) and recent postmenopausal (54.2 ± 2.8 yr) women underwent cardiac magnetic resonance imaging (cMRI) at baseline and after 12 wk of high-intensity aerobic training. Measurements included LV morphology, systolic function, and myocardial perfusion at rest and during an adenosine stress test. At baseline, resting myocardial perfusion was lower in the postmenopausal than the premenopausal group (77 ± 3 vs. 89 ± 3 ml·100 g-1·min-1; P = 0.01), while adenosine-induced myocardial perfusion was not different (P = 0.81). After exercise training, resting myocardial perfusion was lower in both groups (66 ± 2; P = 0.002 vs. 81 ± 3 ml·100 g-1·min-1; P = 0.03). The adenosine-induced change in myocardial perfusion was lower in the groups combined (by 402 ± 17 ml·100 g-1·min-1; P = 0.02), and the adenosine-induced increase in heart rate was 10 ± 2 beats/min lower (P < 0.0001) in both groups after training. Normalization of myocardial perfusion using an estimate of cardiac work eliminated the differences in perfusion between the premenopausal and postmenopausal groups and the effect of training. Left ventricle mass was higher in both groups (P = 0.03; P = 0.006), whereas LV end-diastolic (P = 0.02) and stroke (P = 0.045) volumes were higher in the postmenopausal group after training. Twelve weeks of exercise training increased left ventricle mass and lowered resting and adenosine-induced myocardial perfusion, an effect that was likely related to cardiac work. The current data also suggest that the early menopausal transition has limited impact on cardiac function and structure. NEW & NOTEWORTHY This study provides for the first time estimates of myocardial perfusion in late premenopausal and recent postmenopausal women before and after a period of intense aerobic training. Resting myocardial perfusion was lower in postmenopausal than premenopausal women. Training lowered myocardial resting and stress perfusion in both groups, an effect that was likely influenced by the lower heart rate.
Collapse
Affiliation(s)
- Jon Egelund
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Michael Nyberg
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Camilla M Mandrup
- Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jawdat Abdulla
- Department of Medicine, Division of Cardiology, Glostrup Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens Bangsbo
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Henrik Bo Wiberg Larsson
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
23
|
Nguyen LS, Coutance G, Salem JE, Ouldamar S, Lebreton G, Combes A, Amour J, Laali M, Leprince P, Varnous S. Effect of recipient gender and donor-specific antibodies on antibody-mediated rejection after heart transplantation. Am J Transplant 2019; 19:1160-1167. [PMID: 30286278 DOI: 10.1111/ajt.15133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 08/21/2018] [Accepted: 09/23/2018] [Indexed: 01/25/2023]
Abstract
Gender-difference regarding antibody-mediated rejection (AMR) after heart transplantation has been described. However, no study accounted for the presence of preformed donor-specific antibodies (pfDSA), a known risk factor of AMR, more common among women than men. In a single-institution 6-year cohort (2010-2015), time to AMR was assessed, comparing men with women by survival analysis with a 1-year death-censored follow-up. All AMRs were biopsy proven. Confounding variables that were accounted for included mean intensity fluorescence (MFI) of pfDSA, recipient age, HLA-, size- and sex-mismatch. 463 patients were included. Overall incidence of AMR was 10.3% at 1 year. After adjusting for confounding variables, independent risk factors of AMR were female recipient gender (adjusted hazard-ratio [adj. HR] = 1.78 [1.06-2.99]), P = .03) and the presence of pfDSA (adj. HR = 3.20 [1.80-5.70], P < .001). This association remained significant when considering pfDSA by their MFI; female recipient gender had an adj. HR = 2.2 (P = .026) and MFI of pfDSA (per 1 MFI-increase) adj. HR = 1.0002 (P < .0001). In this cohort, women were at higher risk of AMR than men and this risk increase was additive to that of pfDSA. These findings may suggest a gender-related difference in the severity of pfDSA.
Collapse
Affiliation(s)
- Lee S Nguyen
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France.,APHP, Pitié-Salpétrière, Sorbonne University, Center of Clinical Investigation, ICAN, Paris, France
| | - Guillaume Coutance
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| | - Joe-Elie Salem
- APHP, Pitié-Salpétrière, Sorbonne University, Center of Clinical Investigation, ICAN, Paris, France.,Department of Medicine, Clinical Pharmacology, Cardio-oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Salima Ouldamar
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| | - Guillaume Lebreton
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| | - Alain Combes
- APHP, Pitié-Salpétrière, Sorbonne University, Intensive Care Medicine Department, ICAN, Paris, France
| | - Julien Amour
- APHP, Pitié-Salpétrière, Sorbonne University, Anesthesiology & Critical Care Medicine Department, Paris, France
| | - Mojgan Laali
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| | - Pascal Leprince
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| | - Shaida Varnous
- APHP, Pitié-Salpétrière, Sorbonne University, Cardiac Surgery Department, Institute of Cardiology, Paris, France
| |
Collapse
|
24
|
Barba I, Andrés M, Picón I, Aguade-Bruix S, Garcia-Dorado D. Sex differences in the 1H NMR metabolic profile of serum in cardiovascular risk patients. Sci Rep 2019; 9:2380. [PMID: 30787362 PMCID: PMC6382944 DOI: 10.1038/s41598-019-38881-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 01/11/2019] [Indexed: 01/11/2023] Open
Abstract
Personalized diagnosis and risk stratification of cardiovascular diseases would allow optimizing therapeutic strategies and lifestyle changes. Metabolomics is a promising technique for personalized diagnosis and prognosis; however, various physiological parameters, including sex, influence the metabolic profile thus hampering its translation to the clinic. Knowledge of the variation in the metabolic profile associated with sex would facilitate metabolomic translation to the clinic. The objective of the present work was to investigate the possible differences in the metabolic 1H NMR profile associated to sex beyond lipoproteins. 1H NMR spectra from whole serum and methanol deproteinized samples from 39 patients (22 males, 17 females) between 55-70 years old with suspected coronary artery disease that underwent a stress test that was considered negative where included. Deproteinized serum could be used to differentiate sex based on higher levels of lactate and glucose in women. Lipoprotein region was the most variable area of the spectra between individuals, but spectra of whole serum were able to differentiate sex based on lipoproteins. There are sex-related differences in the 1H NMR metabolic profile of individuals with suspected cardiovascular disease beyond lipoproteins. These findings may help the translation of metabolomics to the clinic.
Collapse
Affiliation(s)
- Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBER-CV), Barcelona, Spain.
| | - Mireia Andrés
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irene Picón
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Santiago Aguade-Bruix
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Garcia-Dorado
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBER-CV), Barcelona, Spain
| |
Collapse
|
25
|
Dai Q, Likes CE, Luz AL, Mao L, Yeh JS, Wei Z, Kuchibhatla M, Ilkayeva OR, Koves TR, Price TM. A Mitochondrial Progesterone Receptor Increases Cardiac Beta-Oxidation and Remodeling. J Endocr Soc 2019; 3:446-467. [PMID: 30746505 PMCID: PMC6364628 DOI: 10.1210/js.2018-00219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/28/2018] [Indexed: 11/21/2022] Open
Abstract
Progesterone is primarily a pregnancy-related hormone, produced in substantial quantities after ovulation and during gestation. Traditionally known to function via nuclear receptors for transcriptional regulation, there is also evidence of nonnuclear action. A previously identified mitochondrial progesterone receptor (PR-M) increases cellular respiration in cell models. In these studies, we demonstrated that expression of PR-M in rat H9c2 cardiomyocytes resulted in a ligand-dependent increase in oxidative cellular respiration and beta-oxidation. Cardiac expression in a TET-On transgenic mouse resulted in gene expression of myofibril proteins for remodeling and proteins involved in oxidative phosphorylation and fatty acid metabolism. In a model of increased afterload from constant transverse aortic constriction, mice expressing PR-M showed a ligand-dependent preservation of cardiac function. From these observations, we propose that PR-M is responsible for progesterone-induced increases in cellular energy production and cardiac remodeling to meet the physiological demands of pregnancy.
Collapse
Affiliation(s)
- Qunsheng Dai
- Division of Reproductive Endocrinology, Duke University, Durham, North Carolina
| | - Creighton E Likes
- Division of Reproductive Endocrinology, Duke University, Durham, North Carolina
| | - Anthony L Luz
- Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Lan Mao
- Division of Cardiology, Duke University, Durham, North Carolina
| | - Jason S Yeh
- Division of Reproductive Endocrinology, Duke University, Durham, North Carolina
| | - Zhengzheng Wei
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina
| | - Maragatha Kuchibhatla
- Division of Biostatistics and Bioinformatics, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, North Carolina
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Timothy R Koves
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina.,Division of Geriatrics, Duke University, Durham, North Carolina
| | - Thomas M Price
- Division of Reproductive Endocrinology, Duke University, Durham, North Carolina
| |
Collapse
|
26
|
Liu LX, Rowe GC, Yang S, Li J, Damilano F, Chan MC, Lu W, Jang C, Wada S, Morley M, Hesse M, Fleischmann BK, Rabinowitz JD, Das S, Rosenzweig A, Arany Z. PDK4 Inhibits Cardiac Pyruvate Oxidation in Late Pregnancy. Circ Res 2017; 121:1370-1378. [PMID: 28928113 DOI: 10.1161/circresaha.117.311456] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 01/23/2023]
Abstract
RATIONALE Pregnancy profoundly alters maternal physiology. The heart hypertrophies during pregnancy, but its metabolic adaptations, are not well understood. OBJECTIVE To determine the mechanisms underlying cardiac substrate use during pregnancy. METHODS AND RESULTS We use here 13C glucose, 13C lactate, and 13C fatty acid tracing analyses to show that hearts in late pregnant mice increase fatty acid uptake and oxidation into the tricarboxylic acid cycle, while reducing glucose and lactate oxidation. Mitochondrial quantity, morphology, and function do not seem altered. Insulin signaling seems intact, and the abundance and localization of the major fatty acid and glucose transporters, CD36 (cluster of differentiation 36) and GLUT4 (glucose transporter type 4), are also unchanged. Rather, we find that the pregnancy hormone progesterone induces PDK4 (pyruvate dehydrogenase kinase 4) in cardiomyocytes and that elevated PDK4 levels in late pregnancy lead to inhibition of PDH (pyruvate dehydrogenase) and pyruvate flux into the tricarboxylic acid cycle. Blocking PDK4 reverses the metabolic changes seen in hearts in late pregnancy. CONCLUSIONS Taken together, these data indicate that the hormonal environment of late pregnancy promotes metabolic remodeling in the heart at the level of PDH, rather than at the level of insulin signaling.
Collapse
Affiliation(s)
- Laura X Liu
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Glenn C Rowe
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Steven Yang
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Jian Li
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Federico Damilano
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Mun Chun Chan
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Wenyun Lu
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Cholsoon Jang
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Shogo Wada
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Michael Morley
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Michael Hesse
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Bernd K Fleischmann
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Joshua D Rabinowitz
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Saumya Das
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Anthony Rosenzweig
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.)
| | - Zoltan Arany
- From the Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA (L.X.L., F.D.); Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston (L.X.L., F.D., M.C.C., S.D., A.R.); Division of Cardiovascular Disease, University of Alabama at Birmingham (G.C.R.); Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.Y., J.L., S.W., M.M., Z.A.); Lewis-Sigler Institute for Integrative Genomics, Princeton University, NJ (W.L., C.J., J.D.R.); and Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (M.H., B.K.F.).
| |
Collapse
|
27
|
Breedt E, Lacerda L, Essop MF. Trimetazidine therapy for diabetic mouse hearts subjected to ex vivo acute heart failure. PLoS One 2017; 12:e0179509. [PMID: 28632748 PMCID: PMC5478112 DOI: 10.1371/journal.pone.0179509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/30/2017] [Indexed: 11/19/2022] Open
Abstract
Acute heart failure (AHF) is the most common primary diagnosis for hospitalized heart diseases in Africa. As increased fatty acid β-oxidation (FAO) during heart failure triggers detrimental effects on the myocardium, we hypothesized that trimetazidine (TMZ) (partial FAO inhibitor) offers cardioprotection under normal and obese-related diabetic conditions. Hearts were isolated from 12-14-week-old obese male and female diabetic (db/db) mice versus lean non-diabetic littermates (db/+) controls. The Langendorff retrograde isolated heart perfusion system was employed to establish an ex vivo AHF model: a) Stabilization phase-Krebs Henseleit buffer (10 mM glucose) at 100 mmHg (25 min); b) Critical Acute Heart Failure (CAHF) phase-(1.2 mM palmitic acid, 2.5 mM glucose) at 20 mmHg (25 min); and c) Recovery Acute Heart Failure phase (RAHF)-(1.2 mM palmitic acid, 10 mM glucose) at 100 mmHg (25 min). Treated groups received 5 μM TMZ in the perfusate during either the CAHF or RAHF stage for the full duration of each respective phase. Both lean and obese males benefited from TMZ treatment administered during the RAHF phase. Sex differences were observed only in lean groups where the phases of the estrous cycle influenced therapy; only the lean follicular female group responded to TMZ treatment during the CAHF phase. Lean luteal females rather displayed an inherent cardioprotection (without treatments) that was lost with obesity. However, TMZ treatment initiated during RAHF was beneficial for obese luteal females. TMZ treatment triggered significant recovery for male and obese female hearts when administered during RAHF. There were no differences between lean and obese male hearts, while lean females displayed a functional recovery advantage over lean males. Thus TMZ emerges as a worthy therapeutic target to consider for AHF treatment in normal and obese-diabetic individuals (for both sexes), but only when administered during the recovery phase and not during the very acute stages.
Collapse
Affiliation(s)
- Emilene Breedt
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Lydia Lacerda
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M. Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
28
|
Barba I, Miró-Casas E, Torrecilla JL, Pladevall E, Tejedor S, Sebastián-Pérez R, Ruiz-Meana M, Berrendero JR, Cuevas A, García-Dorado D. High-fat diet induces metabolic changes and reduces oxidative stress in female mouse hearts. J Nutr Biochem 2017; 40:187-193. [DOI: 10.1016/j.jnutbio.2016.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/26/2016] [Accepted: 11/14/2016] [Indexed: 12/11/2022]
|
29
|
Bartling B, Al-Robaiy S, Lehnich H, Binder L, Hiebl B, Simm A. Sex-related differences in the wheel-running activity of mice decline with increasing age. Exp Gerontol 2017; 87:139-147. [DOI: 10.1016/j.exger.2016.04.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 01/19/2023]
|
30
|
Puoti F, Ricci A, Nanni-Costa A, Ricciardi W, Malorni W, Ortona E. Organ transplantation and gender differences: a paradigmatic example of intertwining between biological and sociocultural determinants. Biol Sex Differ 2016; 7:35. [PMID: 27471591 PMCID: PMC4964018 DOI: 10.1186/s13293-016-0088-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/19/2016] [Indexed: 11/20/2022] Open
Abstract
Organ transplantation, e.g., of the heart, liver, or kidney, is nowadays a routine strategy to counteract several lethal human pathologies. From literature data and from data obtained in Italy, a striking scenario appears well evident: women are more often donors than recipients. On the other hand, recipients of organs are mainly males, probably reflecting a gender bias in the incidence of transplant-related pathologies. The impact of sex mismatch on transplant outcome remains debated, even though donor-recipient sex mismatch, due to biological matters, appears undesirable in female recipients. In our opinion, the analysis of how sex and gender can interact and affect grafting success could represent a mandatory task for the management of organ transplantation.
Collapse
Affiliation(s)
- Francesca Puoti
- National Transplant Center (CNT), Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Ricci
- National Transplant Center (CNT), Istituto Superiore di Sanità, Rome, Italy
| | | | - Walter Ricciardi
- National Transplant Center (CNT), Istituto Superiore di Sanità, Rome, Italy ; Section of Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Walter Malorni
- Section of Gender Medicine, Istituto Superiore di Sanità, Rome, Italy ; Department of Therapeutic Research and Medicine Evaluation, Section of Cell Aging and Degeneration, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Elena Ortona
- Section of Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
31
|
Testosterone Replacement Modulates Cardiac Metabolic Remodeling after Myocardial Infarction by Upregulating PPARα. PPAR Res 2016; 2016:4518754. [PMID: 27413362 PMCID: PMC4927959 DOI: 10.1155/2016/4518754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/17/2016] [Accepted: 05/24/2016] [Indexed: 01/04/2023] Open
Abstract
Despite the importance of testosterone as a metabolic hormone, its effects on myocardial metabolism in the ischemic heart remain unclear. Myocardial ischemia leads to metabolic remodeling, ultimately resulting in ATP deficiency and cardiac dysfunction. In the present study, the effects of testosterone replacement on the ischemic heart were assessed in a castrated rat myocardial infarction model established by ligating the left anterior descending coronary artery 2 weeks after castration. The results of real-time PCR and Western blot analyses showed that peroxisome proliferator-activated receptor α (PPARα) decreased in the ischemic myocardium of castrated rats, compared with the sham-castration group, and the mRNA expression of genes involved in fatty acid metabolism (the fatty acid translocase CD36, carnitine palmitoyltransferase I, and medium-chain acyl-CoA dehydrogenase) and glucose transporter-4 also decreased. A decline in ATP levels in the castrated rats was accompanied by increased cardiomyocyte apoptosis and fibrosis and impaired cardiac function, compared with the sham-castration group, and these detrimental effects were reversed by testosterone replacement. Taken together, our findings suggest that testosterone can modulate myocardial metabolic remodeling by upregulating PPARα after myocardial infarction, exerting a protective effect on cardiac function.
Collapse
|
32
|
Chen J, Young ME, Chatham JC, Crossman DK, Dell'Italia LJ, Shalev A. TXNIP regulates myocardial fatty acid oxidation via miR-33a signaling. Am J Physiol Heart Circ Physiol 2016; 311:H64-75. [PMID: 27199118 DOI: 10.1152/ajpheart.00151.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023]
Abstract
Myocardial fatty acid β-oxidation is critical for the maintenance of energy homeostasis and contractile function in the heart, but its regulation is still not fully understood. While thioredoxin-interacting protein (TXNIP) has recently been implicated in cardiac metabolism and mitochondrial function, its effects on β-oxidation have remained unexplored. Using a new cardiomyocyte-specific TXNIP knockout mouse and working heart perfusion studies, as well as loss- and gain-of-function experiments in rat H9C2 and human AC16 cardiomyocytes, we discovered that TXNIP deficiency promotes myocardial β-oxidation via signaling through a specific microRNA, miR-33a. TXNIP deficiency leads to increased binding of nuclear factor Y (NFYA) to the sterol regulatory element binding protein 2 (SREBP2) promoter, resulting in transcriptional inhibition of SREBP2 and its intronic miR-33a. This allows for increased translation of the miR-33a target genes and β-oxidation-promoting enzymes, carnitine octanoyl transferase (CROT), carnitine palmitoyl transferase 1 (CPT1), hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase-β (HADHB), and AMPKα and is associated with an increase in phospho-AMPKα and phosphorylation/inactivation of acetyl-CoA-carboxylase. Thus, we have identified a novel TXNIP-NFYA-SREBP2/miR-33a-AMPKα/CROT/CPT1/HADHB pathway that is conserved in mouse, rat, and human cardiomyocytes and regulates myocardial β-oxidation.
Collapse
Affiliation(s)
- Junqin Chen
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Martin E Young
- Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - John C Chatham
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - David K Crossman
- Bioinformatics; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Louis J Dell'Italia
- Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anath Shalev
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
33
|
Goldenberg JR, Wang X, Lewandowski ED. Acyl CoA synthetase-1 links facilitated long chain fatty acid uptake to intracellular metabolic trafficking differently in hearts of male versus female mice. J Mol Cell Cardiol 2016; 94:1-9. [PMID: 26995156 DOI: 10.1016/j.yjmcc.2016.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/18/2022]
Abstract
RATIONALE Acyl CoA synthetase-1 (ACSL1) is localized at intracellular membranes, notably the mitochondrial membrane. ACSL1 and female sex are suggested to indirectly facilitate lipid availability to the heart and other organs. However, such mechanisms in intact, functioning myocardium remain unexplored, and roles of ACSL1 and sex in the uptake and trafficking of fats are poorly understood. OBJECTIVE To determine the potential for ACSL1 and sex-dependent differences in metabolic trapping and trafficking effects of long-chain fatty acids (LCFA) within cardiomyocytes of intact hearts. METHODS AND RESULTS (13)C NMR of intact, beating mouse hearts, supplied (13)C palmitate, revealed 44% faster trans-sarcolemmal uptake of LCFA in male hearts overexpressing ACSL1 (MHC-ACSL1) than in non-transgenic (NTG) males (p<0.05). Acyl CoA content was elevated by ACSL1 overexpression, 404% in males and 164% in female, relative to NTG. Despite similar ACSL1 content, NTG females displayed faster LCFA uptake kinetics compared to NTG males, which was reversed by ovariectomy. NTG female LCFA uptake rates were similar to those in ACSL1 males and ACSL1 females. ACSL1 and female sex hormones both accelerated LCFA uptake without affecting triglyceride content or turnover. ACSL1 hearts contained elevated ceramide, particularly C22 ceramide in both sexes and specifically, C24 in males. ACSL1 also induced lower content of fatty acid transporter-6 (FATP6) indicating cooperative regulation with ACSL1. Surprisingly, ACSL1 overexpression did not increase mitochondrial oxidation of exogenous palmitate, which actually dropped in female ACSL1 hearts. CONCLUSIONS ACSL1-mediated metabolic trapping of exogenous LCFA accelerates LCFA uptake rates, albeit to a lesser extent in females, which distinctly affects LCFA trafficking to acyl intermediates but not triglyceride storage or mitochondrial oxidation and is affected by female sex hormones.
Collapse
Affiliation(s)
- Joseph R Goldenberg
- Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago, 909 South Wolcott Avenue, Chicago, IL 60612, USA; Department of Physiology and Biophysics, University of Illinois College of Medicine at Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - Xuerong Wang
- Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago, 909 South Wolcott Avenue, Chicago, IL 60612, USA
| | - E Douglas Lewandowski
- Center for Cardiovascular Research, University of Illinois College of Medicine at Chicago, 909 South Wolcott Avenue, Chicago, IL 60612, USA; Department of Physiology and Biophysics, University of Illinois College of Medicine at Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, USA; Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA.
| |
Collapse
|
34
|
Ruiz M, Gélinas R, Vaillant F, Lauzier B, Des Rosiers C. Metabolic Tracing Using Stable Isotope-Labeled Substrates and Mass Spectrometry in the Perfused Mouse Heart. Methods Enzymol 2015; 561:107-47. [PMID: 26358903 DOI: 10.1016/bs.mie.2015.06.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There has been a resurgence of interest for the field of cardiac metabolism catalyzed by evidence demonstrating a role of metabolic dysregulation in the pathogenesis of heart disease as well as the increased need for new therapeutic targets for patients with these diseases. In this regard, measuring substrate fluxes is critical in providing insight into the dynamics of cellular metabolism and in delineating the regulation of metabolite production and utilization. This chapter provides a comprehensive description of concepts, guidelines, and tips to assess metabolic fluxes relevant to energy substrate metabolism using (13)C-labeled substrates and (13)C-isotopomer analysis by gas chromatography-mass spectrometry (GC-MS), and the ex vivo working heart as study model. The focus will be on the mouse and on flux parameters, which are commonly assessed in the field, namely, those relevant to substrate selection for energy metabolism, specifically the relative contribution of carbohydrate (glucose, lactate, and pyruvate) and fatty acid oxidation to acetyl-CoA formation for citrate synthesis, glycolysis, as well as anaplerosis. We provide detailed procedures for the heart isolation and perfusion in the working mode as well as for sample processing for metabolite extraction and analysis by GC-MS and subsequent data processing for calculation of metabolic flux parameters. Finally, we address practical considerations and discuss additional applications and future challenges.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Roselle Gélinas
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Fanny Vaillant
- IHU Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux, Université de Bordeaux, Bordeaux, France; Inserm U1045 Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | | | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
|
36
|
Bönner F, Janzarik N, Jacoby C, Spieker M, Schnackenburg B, Range F, Butzbach B, Haberkorn S, Westenfeld R, Neizel-Wittke M, Flögel U, Kelm M. Myocardial T2 mapping reveals age- and sex-related differences in volunteers. J Cardiovasc Magn Reson 2015; 17:9. [PMID: 25656484 PMCID: PMC4318191 DOI: 10.1186/s12968-015-0118-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 01/13/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND T2 mapping indicates to be a sensitive method for detection of tissue oedema hidden beyond the detection limits of T2-weighted Cardiovascular Magnetic Resonance (CMR). However, due to variability of baseline T2 values in volunteers, reference values need to be defined. Therefore, the aim of the study was to investigate the effects of age and sex on quantitative T2 mapping with a turbo gradient-spin-echo (GRASE) sequence at 1.5 T. For that reason, we studied sensitivity issues as well as technical and biological effects on GRASE-derived myocardial T2 maps. Furthermore, intra- and interobserver variability were calculated using data from a large volunteer group. METHODS GRASE-derived multiecho images were analysed using dedicated software. After sequence optimization, validation and sensitivity measurements were performed in muscle phantoms ex vivo and in vivo. The optimized parameters were used to analyse CMR images of 74 volunteers of mixed sex and a wide range of age with typical prevalence of hypertension and diabetes. Myocardial T2 values were analysed globally and according to the 17 segment model. Strain-encoded (SENC) imaging was additionally performed to investigate possible effects of myocardial strain on global or segmental T2 values. RESULTS Ex vivo studies in muscle phantoms showed, that GRASE-derived T2 values were comparable to those acquired by a standard multiecho spinecho sequence but faster by a factor of 6. Besides that, T2 values reflected tissue water content. The in vivo measurements in volunteers revealed intra- and interobserver correlations with R2=0.91 and R2=0.94 as well as a coefficients of variation of 2.4% and 2.2%, respectively. While global T2 time significantly decreased towards the heart basis, female volunteers had significant higher T2 time irrespective of myocardial region. We found no correlation of segmental T2 values with maximal systolic, diastolic strain or heart rate. Interestingly, volunteers´ age was significantly correlated to T2 time while that was not the case for other coincident cardiovascular risk factors. CONCLUSION GRASE-derived T2 maps are highly reproducible. However, female sex and aging with typical prevalence of hypertension and diabetes were accompanied by increased myocardial T2 values. Thus, sex and age must be considered as influence factors when using GRASE in a diagnostic manner.
Collapse
Affiliation(s)
- Florian Bönner
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Niko Janzarik
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christoph Jacoby
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
- />Department of Molecular Cardiology, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Maximilian Spieker
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | | | - Felix Range
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Britta Butzbach
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Sebastian Haberkorn
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Ralf Westenfeld
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Mirja Neizel-Wittke
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Ulrich Flögel
- />Department of Molecular Cardiology, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- />CARID (Cardiovascular Research Institute Düsseldorf), Düsseldorf, Germany
| | - Malte Kelm
- />Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University, Medical Faculty, Moorenstraße 5, 40225 Düsseldorf, Germany
- />CARID (Cardiovascular Research Institute Düsseldorf), Düsseldorf, Germany
| |
Collapse
|
37
|
Reiser PJ, Moravec CS. Sex differences in myosin heavy chain isoforms of human failing and nonfailing atria. Am J Physiol Heart Circ Physiol 2014; 307:H265-72. [DOI: 10.1152/ajpheart.00810.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mammalian hearts express two myosin heavy chain (MHC) isoforms, which drive contractions with different kinetics and power-generating ability. The expression of the isoform that is associated with more rapid contraction kinetics and greater power output, MHC-α, is downregulated, with a concurrent increase in the relative amount of the slower isoform, MHC-β, during the progression to experimentally induced or disease-related heart failure. This change in protein expression has been well studied in right and left ventricles in heart failure models and in humans with failure. Relatively little quantitative data exists regarding MHC isoform expression shifts in human failing atria. We previously reported significant increases in the relative amount of MHC-β in the human failing left atrium. The results of that study suggested that there might be a sex-related difference in the level of MHC-β in the left atrium, but the number of female subjects was insufficient for statistical analysis. The objective of this study was to test whether there is, in fact, a sex-related difference in the level of MHC-β in the right and left atria of humans with cardiomyopathy. The results indicate that significant differences exist in atrial MHC isoform expression between men and women who are in failure. The results also revealed an unexpected twofold greater amount of MHC-β in the nonfailing left atrium of women, compared with men. The observed sex-related differences in MHC isoform expression could impact ventricular diastolic filling during normal daily activities, as well as during physiologically stressful events.
Collapse
Affiliation(s)
- Peter J. Reiser
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, Ohio; and
| | | |
Collapse
|
38
|
Nio AQX, Stöhr EJ, Shave R. The female human heart at rest and during exercise: A review. Eur J Sport Sci 2014; 15:286-95. [DOI: 10.1080/17461391.2014.936323] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
Aldehyde dehydrogenase 1A1: friend or foe to female metabolism? Nutrients 2014; 6:950-73. [PMID: 24594504 PMCID: PMC3967171 DOI: 10.3390/nu6030950] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/08/2014] [Accepted: 02/18/2014] [Indexed: 02/08/2023] Open
Abstract
In this review, we summarize recent advances in understanding vitamin A-dependent regulation of sex-specific differences in metabolic diseases, inflammation, and certain cancers. We focus on the characterization of the aldehyde dehydrogenase-1 family of enzymes (ALDH1A1, ALDH1A2, ALDH1A3) that catalyze conversion of retinaldehyde to retinoic acid. Additionally, we propose a “horizontal transfer of signaling” from estrogen to retinoids through the action of ALDH1A1. Although estrogen does not directly influence expression of Aldh1a1, it has the ability to suppress Aldh1a2 and Aldh1a3, thereby establishing a female-specific mechanism for retinoic acid generation in target tissues. ALDH1A1 regulates adipogenesis, abdominal fat formation, glucose tolerance, and suppression of thermogenesis in adipocytes; in B cells, ALDH1A1 plays a protective role by inducing oncogene suppressors Rara and Pparg. Considering the conflicting responses of Aldh1a1 in a multitude of physiological processes, only tissue-specific regulation of Aldh1a1 can result in therapeutic effects. We have shown through successful implantation of tissue-specific Aldh1a1−/− preadipocytes that thermogenesis can be induced in wild-type adipose tissues to resolve diet-induced visceral obesity in females. We will briefly discuss the emerging role of ALDH1A1 in multiple myeloma, the regulation of reproduction, and immune responses, and conclude by discussing the role of ALDH1A1 in future therapeutic applications.
Collapse
|
40
|
Sexual dimorphism of cardiovascular ischemia susceptibility is mediated by heme oxygenase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:521563. [PMID: 24163720 PMCID: PMC3791627 DOI: 10.1155/2013/521563] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 06/25/2013] [Indexed: 01/03/2023]
Abstract
We investigated the gender differences in heme-oxygenase (HO) enzyme, which produces endogenous vascular protective carbon monoxide (CO). We studied (1) the activity and expression of HO enzymes in the left ventricle (LV) and aorta, (2) basal increase in basal blood pressure provoked by arginine vasopressine (AVP) in vivo, (3) the heart perfusion induced by AVP, (4) the ST segment depression provoked by adrenaline and 30 seconds later phentolamine, and (5) the aorta ring contraction induced by AVP in female and male Wistar rats. We found that HO activity and the expression of HO-1 and HO-2 were increased in female rat aorta and LV. We demonstrated that the basal blood pressure and administration of AVP provoked blood pressure response are increased in the males; the female myocardium was less sensitive towards angina. Both differences could be aggravated by the inhibition of HO. The aorta rings were more susceptible towards vasoconstriction by AVP in males; isolated heart perfusion decrease was higher in males. The HO inhibition aggravated the heart perfusion in both sexes. In conclusion, the increased HO activity and expression in females might play a role in the sexual dimorphism of cardiovascular ischemia susceptibility during the reproductive age.
Collapse
|
41
|
Classical estrogen receptors and ERα splice variants in the mouse. PLoS One 2013; 8:e70926. [PMID: 23940668 PMCID: PMC3733772 DOI: 10.1371/journal.pone.0070926] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/24/2013] [Indexed: 12/18/2022] Open
Abstract
Estrogens exert a variety of effects in both reproductive and non-reproductive tissues. With the discovery of ERα splice variants, prior assumptions concerning tissue-specific estrogen signaling need to be re-evaluated. Accordingly, we sought to determine the expression of the classical estrogen receptors and ERα splice variants across reproductive and non-reproductive tissues of male and female mice. Western blotting revealed that the full-length ERα66 was mainly present in female reproductive tissues but was also found in non-reproductive tissues at lower levels. ERα46 was most highly expressed in the heart of both sexes. ERα36 was highly expressed in the kidneys and liver of female mice but not in the kidneys of males. ERβ was most abundant in non-reproductive tissues and in the ovaries. Because the kidney has been reported to be the most estrogenic non-reproductive organ, we sought to elucidate ER renal expression and localization. Immunofluorescence studies revealed ERα66 in the vasculature and the glomerulus. It was also found in the brush border of the proximal tubule and in the cortical collecting duct of female mice. ERα36 was evident in mesangial cells and tubular epithelial cells of both sexes, as well as podocytes of females but not males. ERβ was found primarily in the podocytes in female mice but was also present in the mesangial cells in both sexes. Within the renal cortex, ERα46 and ERα36 were mainly located in the membrane fraction although they were also present in the cytosolic fraction. Given the variability of expression patterns demonstrated herein, identification of the specific estrogen receptors expressed in a tissue is necessary for interpreting estrogenic effects. As this study revealed expression of the ERα splice variants at multiple sites within the kidney, further studies are warranted in order to elucidate the contribution of these receptors to renal estrogen responsiveness.
Collapse
|
42
|
Wattanapermpool J, de Tombe PP, Pak TR. Sex differences in health and disease: brain and heart connections--a special issue. Pflugers Arch 2013; 465:555-6. [PMID: 23588381 DOI: 10.1007/s00424-013-1279-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/03/2013] [Indexed: 11/28/2022]
|
43
|
Gong Y, Xiao H, Li C, Bai J, Cheng X, Jin M, Sun B, Lu Y, Shao Y, Tian H. Elevated t/e2 ratio is associated with an increased risk of cerebrovascular disease in elderly men. PLoS One 2013; 8:e61598. [PMID: 23637864 PMCID: PMC3634802 DOI: 10.1371/journal.pone.0061598] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/11/2013] [Indexed: 11/28/2022] Open
Abstract
Objective To investigate the relationship between sex hormones and the risk of vascular disease in elderly men and to evaluate the advantages and disadvantages of testosterone replacement. Methods A total of 337 men, aged 60 to 91 years, were enrolled in this single-center, cross-sectional study, and their sex hormone levels were assessed. Linear and logistic regression analyses were utilized to compare the sex hormone levels between patients with and without vascular disease. The nonparametric K-sample test was used for inter-group comparisons. Results Aging and abnormal metabolism were both significantly associated with an increased risk of vascular diseases and changes in sex hormone levels. Primary linear and logistic regression analyses showed no significant differences in sex hormone concentrations between patients with and without vascular diseases after adjusting for age. Logistic regression with abnormal metabolism as categorical variable showed that free testosterone (FT) and free estradiol (FE2) had significant relationships with CEVD risk (P<0.05). In further regression with all metabolic continuous variables included, the testosterone/estradiol (T/E2) ratio replaced FT and FE2 (P<0.05). Trend line analyses showed that T/E2 actually had a binomial linear correlation with the risk of cerebrovascular disease; its best protective effect occurred at values of 0.13–0.15, with an OR value extremely close to those of FT and FE2 (0.23 vs. 0.24–0.25). Conclusion T/E2 balance plays a key role in the relationship between sex hormones and the risk of cerebrovascular disease. The balance between T and E2 may be more important than their absolute quantities. Extremely low T/E2 and inappropriately high T/E2 ratio can both harm the brain blood vessels. Careful consideration should be given before beginning testosterone replacement treatment, and supplementing with estrogen seems to be a good way to protect blood vessels of the brain in elderly men.
Collapse
Affiliation(s)
- Yanping Gong
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haiying Xiao
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chunlin Li
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
- * E-mail:
| | - Jie Bai
- Department of Geriatric Clinical Laboratory, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoling Cheng
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Mengmeng Jin
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Boruo Sun
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yanhui Lu
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yinghong Shao
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui Tian
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|