1
|
He Q, Yang J, Chen W, Pan Z, Chen B, Zeng J, Zhang N, Lin Y, Chen C, Xiao J, Li M, Li S, Wang H, Chen P. Biochanin A abrogates osteoclastogenesis in type 2 diabetic osteoporosis via regulating ROS/MAPK signaling pathway based on integrating molecular docking and experimental validation. BMC Complement Med Ther 2024; 24:24. [PMID: 38191438 PMCID: PMC10773052 DOI: 10.1186/s12906-023-04332-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND There are accumulating type 2 diabetes patients who have osteoporosis simultaneously. More effective therapeutic strategies should be discovered. Biochanin A (BCA) has been indicated that can play a role in improving metabolic disorders of type 2 diabetes and preventing osteoporosis. But whether BCA can treat type 2 diabetic osteoporosis has not been studied. PURPOSE To investigate if the BCA can protect against type 2 diabetic osteoporosis and clarify the mechanism. METHODS Micro-CT and histology assays were performed to detect the trabecular bone and analyze the bone histomorphology effect of BCA. CCK-8 assay was performed to detect the toxicity of BCA. TRAcP staining, immunofluorescence and hydroxyapatite resorption assay were used to observe osteoclasts differentiation and resorptive activity. Molecular docking provided evidence about BCA regulating the MAPK axis via prediction by the algorithm. QRT-PCR and Western Blotting were utilized to detect the expression of osteoclastogenesis-related markers and MAPK signaling pathway. RESULTS Accumulation of bone volume after BCA treatment could be found based on the 3D reconstruction. Besides, there were fewer osteoclasts in db/db mice treated with BCA than db/db mice treated with saline. In vitro, we found that BCA hadn't toxicity in osteoclasts precursor, but also inhibited differentiation of osteoclasts. Further, we found that BCA suppresses osteoclastogenesis via ROS/MAPK signaling pathway. CONCLUSION BCA can prevent type 2 diabetic osteoporosis by restricting osteoclast differentiation via ROS/MAPK signaling pathway.
Collapse
Affiliation(s)
- Qi He
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Junzheng Yang
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
- Fifth School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
| | - Weijian Chen
- Fifth School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
| | - Zhaofeng Pan
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Baihao Chen
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Jiaxu Zeng
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Nenling Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, P.R. China
| | - Yuewei Lin
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Chuyi Chen
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Jiacong Xiao
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Miao Li
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Shaocong Li
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China.
| | - Peng Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China.
| |
Collapse
|
2
|
Collins HE. Female cardiovascular biology and resilience in the setting of physiological and pathological stress. Redox Biol 2023; 63:102747. [PMID: 37216702 PMCID: PMC10209889 DOI: 10.1016/j.redox.2023.102747] [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: 03/27/2023] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
For years, females were thought of as smaller men with complex hormonal cycles; as a result, females have been largely excluded from preclinical and clinical research. However, in the last ten years, with the increased focus on sex as a biological variable, it has become clear that this is not the case, and in fact, male and female cardiovascular biology and cardiac stress responses differ substantially. Premenopausal women are protected from cardiovascular diseases, such as myocardial infarction and resultant heart failure, having preserved cardiac function, reduced adverse remodeling, and increased survival. Many underlying biological processes that contribute to ventricular remodeling differ between the sexes, such as cellular metabolism; immune cell responses; cardiac fibrosis and extracellular matrix remodeling; cardiomyocyte dysfunction; and endothelial biology; however, it is unclear how these changes afford protection to the female heart. Although many of these changes are dependent on protection provided by female sex hormones, several of these changes occur independent of sex hormones, suggesting that the nature of these changes is more complex than initially thought. This may be why studies focused on the cardiovascular benefits of hormone replacement therapy in post-menopausal women have provided mixed results. Some of the complexity likely stems from the fact that the cellular composition of the heart is sexually dimorphic and that in the setting of MI, different subpopulations of these cell types are apparent. Despite the documented sex-differences in cardiovascular (patho)physiology, the underlying mechanisms that contribute are largely unknown due to inconsistent findings amongst investigators and, in some cases, lack of rigor in reporting and consideration of sex-dependent variables. Therefore, this review aims to describe current understanding of the sex-dependent differences in the myocardium in response to physiological and pathological stressors, with a focus on the sex-dependent differences that contribute to post-infarction remodeling and resultant functional decline.
Collapse
Affiliation(s)
- Helen E Collins
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, Delia B. Baxter Research Building, University of Louisville, 580 S. Preston S, Louisville, KY 40202, USA.
| |
Collapse
|
3
|
Pires LAS, Babinski MSD, Fonseca Junior A, Manaia JHM, Babinski MA. Aging effects in the extracellular matrix of the clitoris: A scanning electron microscopic analysis. Morphologie 2023; 107:259-263. [PMID: 36707352 DOI: 10.1016/j.morpho.2023.01.001] [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: 10/18/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVE OF THE STUDY The present work aims to observe the clitoris' extracellular matrix in young and old women with a scanning electron microscope. MATERIALS AND METHODS After approval of the local research ethics committee, samples of the clitoris body were obtained from cadavers of women between 20 and 40 old (G1) and from cadavers over the age of 60 (G2). The samples were decellularized with NaOH to maintain the extracellular matrix framework, submitted to silver sputter coating, and observed under a scanning electron microscope. RESULTS The mean age of the cadavers in G1 was 28 years old and 75±6 years old in G2. The groups were composed of 10 cadavers each. It was observed that the collagen was arranged in a disorganized fashion in the samples from the G2 in several regions. There was also a decrease in elastic fibers that anchored the collagen in these samples. The concentration of collagen showed an increase in the older samples in comparison to the G1 samples. Conclusions Female sexual dysfunction is a condition prevalent in a significantly large portion of women and it is more common in elderly women. It is known that the tumescence mechanism requires integrity of the extracellular matrix. The changes observed herein may alter the function of the organ and are similar to observations in studies of men with erectile dysfunction. CONCLUSION Female sexual dysfunction is a condition prevalent in a significantly large portion of women and it is more common in elderly women. It is known that the tumescence mechanism requires integrity of the extracellular matrix. The changes observed herein may alter the function of the organ and are similar to observations in studies of men with erectile dysfunction. .
Collapse
Affiliation(s)
- Lucas Alves Sarmento Pires
- Experimental Morphology Laboratory, Morphology Department, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Medical Sciences Post Graduation Program, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil.
| | - Monique Silva Dias Babinski
- Experimental Morphology Laboratory, Morphology Department, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Medical Sciences Post Graduation Program, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Albino Fonseca Junior
- Experimental Morphology Laboratory, Morphology Department, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Medical Sciences Post Graduation Program, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Jorge Henrique Martins Manaia
- Experimental Morphology Laboratory, Morphology Department, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Medical Sciences Post Graduation Program, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marcio Antonio Babinski
- Experimental Morphology Laboratory, Morphology Department, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Medical Sciences Post Graduation Program, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Lira-Silva E, del Valle Mondragón L, Pérez-Torres I, Posadas-Sánchez R, Roldán Gómez FJ, Posadas-Romero C, Vargas-Barrón J, Pavón N. Possible implication of estrogenic compounds on heart disease in menopausal women. Biomed Pharmacother 2023; 162:114649. [PMID: 37023620 DOI: 10.1016/j.biopha.2023.114649] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Epidemiological studies imply there is a higher risk of cardiovascular disease in menopausal women. Some explanations suggest a lack of estrogens as the cause, but estrogens do not disappear completely and are just transformed into different products called estrogenic degradation metabolites (EDMs). When estrogens are metabolized, reactive oxygen species (ROS) increase, causing DNA damage and increasing oxidative stress. These conditions are associated to neurodegenerative diseases and different types of cancer. However, their effect on the cardiovascular system remains unknown. This paper compares estrogenic metabolite levels in serum from post-menopausal women with cardiovascular risk (CAC>1) and with establish cardiovascular disease (CVD), against levels in healthy women (Ctrl). Sample sera were obtained from the Genetics of Atherosclerotic Disease (GEA) Mexican Study. Serum levels of eleven estrogenic metabolites were quantified by High performance liquid chromatography (HPLC) and oxidative stress markers such as ROS, lipoperoxidation levels (TBARS), total antioxidant capacity (TAC), super oxide dismutase activity (SOD) and cytokine levels were evaluated. 8-hydroxy-2-deoxyguanosine (8-OHdG) was also determined as a marker of nuclear damage.There were significant differences between serum levels of some EDMs in CAC> 1 and CVD vs. serum levels in Ctrl women. Results also revealed an increase in oxidative stress and a diminished capacity to manage oxidative stress. These findings provide an overview, and suggest that some estrogenic metabolites may be associated with an increased risk of CVD in menopausal women. However, additional studies are needed to evaluate the impact of these EDMs directly on cardiovascular function.
Collapse
|
5
|
Akintoye E, Saijo Y, Braghieri L, Badwan O, Patel H, Dabbagh MM, El Dahdah J, Jellis CL, Desai MY, Rodriguez LL, Grimm RA, Griffin BP, Popović ZB. Impact of Age and Sex on Left Ventricular Remodeling in Patients With Aortic Regurgitation. J Am Coll Cardiol 2023; 81:1474-1487. [PMID: 37045517 PMCID: PMC9982944 DOI: 10.1016/j.jacc.2023.02.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Current guidelines for aortic regurgitation (AR) recommend the same linear left ventricular (LV) dimension for intervention regardless of age and sex. OBJECTIVES The purpose of this study was to evaluate the impact of age and sex on the degree of LV remodeling and outcomes. METHODS We included consecutive patients with severe AR who were serially monitored by echocardiogram between 2010 and 2016. The 2 main endpoints were as follows: 1) LV end-systolic volume indexed to body surface area (LVESVi) and LV end-diastolic volume indexed to body surface area; and 2) adverse events (AE). We evaluated the longitudinal rate of LV remodeling and determined the association between LV volume and AE by age and sex. RESULTS A total of 525 adult patients (26% women) with a median echocardiogram follow-up of 2.0 years (IQR: 1.0-3.6 years) were included. At baseline, older patients (age ≥60 years) had smaller LV volumes compared with younger patients (age <60 years), eg, the mean LVESVi was 27.3 mL/m2 vs 32.3 mL/m2, respectively. Similarly, women had smaller LV volumes compared with men (mean LVESVi was 23.3 mL/m2 vs 32.4 mL/m2). On serial evaluation, older patients and women maintained smaller LV volumes compared with younger patients and men, respectively. There were 210 (40%) AE during follow-up. The optimal discriminatory threshold for AE varies by age and sex, eg, the LVESVi threshold was highest for young men (50 mL/m2), intermediate for older men (35 mL/m2), and lowest for women (27 mL/m2). CONCLUSIONS On serial evaluation, older patients and women with chronic AR maintained smaller LV volumes than younger patients and men, respectively, and develop AE at lower LV volumes.
Collapse
Affiliation(s)
- Emmanuel Akintoye
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA. https://twitter.com/eakintoyeMD
| | - Yoshihito Saijo
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lorenzo Braghieri
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Osamah Badwan
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hardik Patel
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - M Marwan Dabbagh
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joseph El Dahdah
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christine L Jellis
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA. https://twitter.com/ChrisJellisMD
| | - Milind Y Desai
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA. https://twitter.com/DesaiMilindY
| | - L Leonardo Rodriguez
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard A Grimm
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brian P Griffin
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA. https://twitter.com/BrianGriffinMD
| | - Zoran B Popović
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
6
|
Patel HP, Thakkar S, Mehta N, Faisaluddin M, Munshi RF, Kumar A, Khan SU, Parikh R, DeSimone CV, Sharma G, Deshmukh A, Nasir K, Ganatra S, Dani SS. Racial disparities in ventricular tachycardia in young adults: analysis of national trends. J Interv Card Electrophysiol 2023; 66:193-202. [PMID: 35947319 DOI: 10.1007/s10840-022-01335-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND In the last two decades, risk factors, prevalence, and mortality due to coronary artery disease in young adults are on the rise. We sought to assess the prevalence, trends, and economic burden of ventricular tachycardia (VT) hospitalizations in young adults (< 45 years), further stratified by race and gender. METHODS The Nationwide Inpatient Sample was explored for hospitalizations with VT in patients (< 45 years) between 2005 and 2018 and divided among 3 groups of the quadrennial period using validated International Classification of Diseases (ICD) 9th and 10th revision Clinical Modification (CM) codes. The Pearson chi-square test and Wilcoxon rank-sum were used for categorical and continuous variables, respectively. We assessed the temporal trends of mortality in VT hospitalizations and trends of VT hospitalization stratified by age, sex, and race by using Joinpoint regression analysis. The primary outcome was in-hospital mortality trends. Secondary outcomes were trends of hospital stay in days, cost of care in US dollars, cardiac arrest, and discharge disposition. RESULTS Out of 5,156,326 patients admitted with VT between 2005 and 2018, 309,636 were young adults. Among them, 102,433 were admitted between 2005 and 2009 (mean age 36.1 ± 6.99; 61% male, 58.5% White), 109,591 between 2010 and 2014 (mean age 35.5 ± 7.16; 59% male, 54.2% White), and 97,495 between 2015 and 2018 (mean age 35.4 ± 7.00; 60% male, 52.3% White) (p < 0.07). In the young adults with VT, all-cause mortality was 7.37% from 2005 to 2009, 7.85% from 2010 to 2014 (6.5% relative increase from 2005 to 2009), and 8.98% from 2015 to 2018 (relative increase of 14.4% from 2010 to 2014) (p < 0.0001). Similarly, risk of cardiac arrest was on the rise (6.15% from 2005 to 2009 to 7.77% in 2010-2014 and 9.97% in 2015-2018). Inflation-adjusted cost increased over the years [$12,177 in 2005-2009; $13,249 in 2010-2014; $15,807 in 2015-2018; p < 0.0001)]. CONCLUSIONS VT hospitalizations and related all-cause mortality, and healthcare utilization costs in young adults are on the rise in the study period. Hospitalization burden related to VT and poor outcomes were more notable for Black adults. Further studies are required for targeted screening and preventative measures in young adults.
Collapse
Affiliation(s)
- Harsh P Patel
- Department of Cardiology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Samarthkumar Thakkar
- Department of Cardiovascular Medicine, Houston Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Nishaki Mehta
- Department of Cardiovascular Medicine, Oakland University William Beaumont School of Medicine, Beaumont Hospital Royal Oak, Royal Oak, MI, USA
| | | | - Rezwan F Munshi
- Department of Medicine, Nassau University Medical Center, East Meadow, NY, USA
| | - Ashish Kumar
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Safi U Khan
- Department of Cardiovascular Medicine, Houston Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Rohan Parikh
- Department of Cardiology, Lahey Hospital & Medical Center, 41 Burlington Mall Road, Burlington, MA, 01805, USA
| | | | - Garima Sharma
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhishek Deshmukh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Khurram Nasir
- Department of Cardiovascular Medicine, Houston Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Sarju Ganatra
- Department of Cardiology, Lahey Hospital & Medical Center, 41 Burlington Mall Road, Burlington, MA, 01805, USA
| | - Sourbha S Dani
- Department of Cardiology, Lahey Hospital & Medical Center, 41 Burlington Mall Road, Burlington, MA, 01805, USA.
| |
Collapse
|
7
|
Kaplan A, Abidi E, Diab R, Ghali R, Al-Awassi H, Booz GW, Zouein FA. Sex differences in cardiac remodeling post myocardial infarction with acute cigarette smoking. Biol Sex Differ 2022; 13:36. [PMID: 35799275 PMCID: PMC9264586 DOI: 10.1186/s13293-022-00446-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Whether cigarette smoking affects the heart post-myocardial infarction (MI) in a sex-dependent way remains controversial. Using a mouse model, we investigated cardiac remodeling under the influence of acute cigarette smoke (CS) exposure following ischemic injury in both sexes. Methods Ten cigarettes were smoked twice daily for 2 weeks followed by MI and then 1 additional week post permanent LAD ligation. Cardiac function, histology, and infarct size were assessed, and inflammatory markers quantified by RT–PCR. Statistical comparisons were performed using an unpaired t test or ANOVA followed by Tukey post hoc test. Results We observed that cigarette smoking exacerbated both left and right ventricular remodeling only in males at an early stage of post-MI. Females did not display a significant structural and/or functional alteration within 7 days of cardiac remodeling post-MI upon CS exposure. Worsened right ventricular remodeling in males was independent of pulmonary congestion. CS-exposed males exhibited enhanced increases in left ventricular end systolic and diastolic volumes, as well as reductions in ejection fraction and fractional area changes of left ventricular base. At day 7, infarct size was increased by cigarette smoking in males only, which was accompanied by enhanced collagen deposition in both the infarcted and peri-infarcted areas. Both IL-6 and TNF-α mRNA expression significantly increased in CS-exposed MI male group only at day 7 post-MI suggestive of prolonged inflammation. Conclusions These findings indicate that CS exposure worsens the progression of cardiac remodeling post-MI in male sex in a significant manner compared to female sex at least at early stages.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon.,Department of Cardiology, Kemer Public Hospital, Hastane Cd. No: 9, 07980, Kemer, Antalya, Turkey.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Emna Abidi
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon.,Department of Pharmacy, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Reine Diab
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Rana Ghali
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Hiam Al-Awassi
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon.,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, 39216-4500, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Medical Center, American University of Beirut, Faculty of Medicine, Riad El-Solh, Beirut, 1107 2020, Lebanon. .,The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon. .,Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, 39216-4500, USA. .,Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, Paris, France.
| |
Collapse
|
8
|
Vafashoar F, Mousavizadeh K, Poormoghim H, Haghighi A, Pashangzadeh S, Mojtabavi N. Progesterone Aggravates Lung Fibrosis in a Mouse Model of Systemic Sclerosis. Front Immunol 2021; 12:742227. [PMID: 34912332 PMCID: PMC8667310 DOI: 10.3389/fimmu.2021.742227] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
Background Gender-related factors have explained the higher prevalence of autoimmune diseases in women. Sex hormones play a key role in the immune system and parenchymal cells function; therefore, these hormones can be important in the pathogenesis of autoimmune diseases as a risk or beneficial factor. Lung fibrosis is the main cause of mortality in systemic sclerosis, a female predominant autoimmune disease. The objective of this study was to examine the effect of progesterone on lung fibrosis in a mouse model of systemic sclerosis. Methods Mice with bleomycin-induced lung fibrosis treated with progesterone subcutaneously for 21 and 28 days. Blood was collected for hormone and cytokine measurement at the end of treatment then, skin and lung tissues were harvested for histological assessment, gene expression, cytokine, hydroxyproline, and gelatinase measurement. Results Trichrome staining and hydroxyproline measurements showed that progesterone treatment increased the content of collagen in fibrotic and normal lung tissues. Progesterone increased α-SMA (P < 0.01), TGF- β (P < 0.05) and decreased MMP9 (P < 0.05) in fibrotic lung tissues. Also progesterone treatment decreased the gene expression of Col1a2 (P <0.05), Ctgf (P <01), End1 (0.001) in bleomycin- injured lung tissues. The serum level of TNF-α was decreased, but the serum level of cortisol was increased by progesterone treatment in fibrotic mice (P< 0.05). Conclusion Our results showed that progesterone aggravates lung fibrosis in a mouse model of systemic sclerosis.
Collapse
Affiliation(s)
- Fatemeh Vafashoar
- Institute of Immunology and Infectious Disease, Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Kazem Mousavizadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Poormoghim
- Scleroderma Study Group, Firuzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Haghighi
- Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Salar Pashangzadeh
- Institute of Immunology and Infectious Disease, Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Institute of Immunology and Infectious Disease, Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Adekunle AO, Adzika GK, Mprah R, Ndzie Noah ML, Adu-Amankwaah J, Rizvi R, Akhter N, Sun H. Predominance of Heart Failure With Preserved Ejection Fraction in Postmenopausal Women: Intra- and Extra-Cardiomyocyte Maladaptive Alterations Scaffolded by Estrogen Deficiency. Front Cell Dev Biol 2021; 9:685996. [PMID: 34660569 PMCID: PMC8511782 DOI: 10.3389/fcell.2021.685996] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains a public health concern as it is associated with high morbidity and death rates. In particular, heart failure with preserved ejection fraction (HFpEF) represents the dominant (>50%) form of HF and mostly occurring among postmenopausal women. Hence, the initiation and progression of the left ventricular diastolic dysfunctions (LVDD) (a typically clinical manifestation of HFpEF) in postmenopausal women have been attributed to estrogen deficiency and the loss of its residue cardioprotective effects. In this review, from a pathophysiological and immunological standpoint, we discuss the probable multiple pathomechanisms resulting in HFpEF, which are facilitated by estrogen deficiency. The initial discussions recap estrogen and estrogen receptors (ERs) and β-adrenergic receptors (βARs) signaling under physiological/pathological states to facilitate cardiac function/dysfunction, respectively. By reconciling these prior discussions, attempts were made to explain how the loss of estrogen facilitates the disruptions both ERs and βARs-mediated signaling responsible for; the modulation of intra-cardiomyocyte calcium homeostasis, maintenance of cardiomyocyte cytoskeletal and extracellular matrix, the adaptive regulation of coronary microvascular endothelial functions and myocardial inflammatory responses. By scaffolding the disruption of these crucial intra- and extra-cardiomyocyte physiological functions, estrogen deficiency has been demonstrated to cause LVDD and increase the incidence of HFpEF in postmenopausal women. Finally, updates on the advancements in treatment interventions for the prevention of HFpEF were highlighted.
Collapse
Affiliation(s)
| | | | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | | | | | - Nazma Akhter
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Abecasis J, Gomes Pinto D, Ramos S, Masci PG, Cardim N, Gil V, Félix A. Left Ventricular Remodeling in Degenerative Aortic Valve Stenosis. Curr Probl Cardiol 2021; 46:100801. [PMID: 33588124 DOI: 10.1016/j.cpcardiol.2021.100801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 01/15/2023]
Abstract
Aortic stenosis was once considered a pure isolated valve obstacle challenging left ventricle driving force of contraction and flow generation. Left ventricular (LV) adaptation was merely interpreted as a uniform hypertrophic response to increased afterload. However, in these last 2 decades cardiac imaging research and some histopathology correlation studies brought insight towards the complex interaction between the vasculature, the valve and the myocardium. Verily, LV remodeling in this setting is a complex multidetermined process that goes further beyond myocardial hypertrophy. Ultrastructural changes involving both diffuse and replacement fibrosis of the myocardium take part and might explain the transition of clinical phenotypes with distinct prognosis, from compensated hypertrophy to LV maladaptive dysfunction and heart failure. Presently, the combined appropriate use of echocardiography and cardiac magnetic resonance may better assess the global LV afterload, hypertrophy and geometric remodeling, global and regional LV function, beyond ejection fraction, and structural changes that include the fibrotic burden of the myocardium. As a whole these may not only better stratify individual risk of disease progression but also identify patients benefiting from earlier valve intervention. In this paper, we review the maladaptive response of the LV to chronic pressure overload, describing the different signaling pathways and mechanisms that underly both hypertrophy and remodeling. Histomorphology changes in this setting are described and we try to make sense of the use of new imaging tools for LV characterization.
Collapse
Affiliation(s)
- João Abecasis
- Nova Medical School, Lisboa, Portugal; Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal; Cardiology Department, Hospital dos Lusíadas, Lisboa, Portugal.
| | - Daniel Gomes Pinto
- Nova Medical School, Lisboa, Portugal; Pathology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | - Sância Ramos
- Pathology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal; Faculdade Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | | | - Nuno Cardim
- Nova Medical School, Lisboa, Portugal; Hospital da Luz, Lisboa, Portugal
| | - Victor Gil
- Cardiology Department, Hospital dos Lusíadas, Lisboa, Portugal; Faculdade de Medicina de Lisboa, Portugal
| | - Ana Félix
- Nova Medical School, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| |
Collapse
|
11
|
Brady TM, Roem J, Cox C, Schneider MF, Wilson AC, Furth SL, Warady BA, Mitsnefes M. Adiposity, Sex, and Cardiovascular Disease Risk in Children With CKD: A Longitudinal Study of Youth Enrolled in the Chronic Kidney Disease in Children (CKiD) Study. Am J Kidney Dis 2020; 76:166-173. [PMID: 32389356 PMCID: PMC7387195 DOI: 10.1053/j.ajkd.2020.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022]
Abstract
RATIONALE & OBJECTIVE Traditional and nontraditional cardiovascular disease risk factors are highly prevalent in children with chronic kidney disease (CKD). We examined the longitudinal association of adiposity with cardiac damage among children with CKD and explored whether this association was modified by sex. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS Children with mild-to-moderate CKD enrolled in the Chronic Kidney Disease in Children (CKiD) Study at 49 pediatric nephrology centers across North America. EXPOSURE Age- and sex-specific body mass index (BMI) z score. OUTCOME Age- and sex-specific left ventricular mass index (LVMI) z score and left ventricular hypertrophy (LVH). ANALYTICAL APPROACH Longitudinal analyses using mixed-effects models to estimate sex-specific associations of BMI z scores with LVMI z score and with LVH, accounting for repeated measurements over time. RESULTS Among 725 children with 2,829 person-years of follow-up, median age was 11.0 years and median estimated glomerular filtration rate was 52.6mL/min/1.73m2. Nearly one-third of both boys and girls were overweight or obese, median LVMI z score was 0.18 (IQR: -0.67, 1.08), and 11% had LVH. Greater BMI z scores were independently associated with greater LVMI z scores and greater odds of LVH. For each 1-unit higher BMI z score, LVMI z score was 0.24 (95% CI, 0.17-0.31) higher in boys and 0.38 (95% CI, 0.29-0.47) higher in girls (Pinteraction = 0.01). For each 1-unit higher BMI z score, the odds of LVH was 1.5-fold (95% CI, 1.1-2.1) higher in boys and 3.1-fold (95% CI, 1.8-4.4) higher in girls (Pinteraction = 0.005). LIMITATIONS Not all children had repeated measurements. LVH is a surrogate and not a hard cardiac outcome. The observational design limits causal inference. CONCLUSIONS In children, adiposity is independently associated with the markers of cardiac damage, LVMI z score and LVH. This association is stronger among girls than boys. Pediatric overweight and obesity may therefore have a substantial impact on cardiovascular risk among children with CKD.
Collapse
Affiliation(s)
- Tammy M Brady
- Division of Pediatric Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Jennifer Roem
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Christopher Cox
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Michael F Schneider
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Amy C Wilson
- J.W. Riley Hospital for Children, Indianapolis, IN
| | - Susan L Furth
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Bradley A Warady
- Division of Nephrology, Children's Mercy Hospital, Kansas City, MO
| | - Mark Mitsnefes
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
12
|
Is Cardiac Diastolic Dysfunction a Part of Post-Menopausal Syndrome? JACC-HEART FAILURE 2020; 7:192-203. [PMID: 30819374 DOI: 10.1016/j.jchf.2018.12.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 11/22/2022]
Abstract
Post-menopausal women exhibit an exponential increase in the incidence of heart failure with preserved ejection fraction compared with men of the same age, which indicates a potential role of hormonal changes in subclinical and clinical diastolic dysfunction. This paper reviews the preclinical evidence that demonstrates the involvement of estrogen in many regulatory molecular pathways of cardiac diastolic function and the clinical data that investigates the effect of estrogen on diastolic function in post-menopausal women. Published reports show that estrogen deficiency influences both early diastolic relaxation via calcium homeostasis and the late diastolic compliance associated with cardiac hypertrophy and fibrosis. Because of the high risk of diastolic dysfunction and heart failure with preserved ejection fraction in post-menopausal women and the positive effects of estrogen on preserving cardiac function, further clinical studies are needed to clarify the role of endogenous estrogen or hormone replacement in mitigating the onset and progression of heart failure with preserved ejection fraction in women.
Collapse
|
13
|
Maione AS, Pilato CA, Casella M, Gasperetti A, Stadiotti I, Pompilio G, Sommariva E. Fibrosis in Arrhythmogenic Cardiomyopathy: The Phantom Thread in the Fibro-Adipose Tissue. Front Physiol 2020; 11:279. [PMID: 32317983 PMCID: PMC7147329 DOI: 10.3389/fphys.2020.00279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disorder, predisposing to malignant ventricular arrhythmias leading to sudden cardiac death, particularly in young and athletic patients. Pathological features include a progressive loss of myocardium with fibrous or fibro-fatty substitution. During the last few decades, different clinical aspects of ACM have been well investigated but still little is known about the molecular mechanisms that underlie ACM pathogenesis, leading to these phenotypes. In about 50% of ACM patients, a genetic mutation, predominantly in genes that encode for desmosomal proteins, has been identified. However, the mutation-associated mechanisms, causing the observed cardiac phenotype are not always clear. Until now, the attention has been principally focused on the study of molecular mechanisms that lead to a prominent myocardium adipose substitution, an uncommon marker for a cardiac disease, thus often recognized as hallmark of ACM. Nonetheless, based on Task Force Criteria for the diagnosis of ACM, cardiomyocytes death associated with fibrous replacement of the ventricular free wall must be considered the main tissue feature in ACM patients. For this reason, it urges to investigate ACM cardiac fibrosis. In this review, we give an overview on the cellular effectors, possible triggers, and molecular mechanisms that could be responsible for the ventricular fibrotic remodeling in ACM patients.
Collapse
Affiliation(s)
- Angela Serena Maione
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Chiara Assunta Pilato
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michela Casella
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Alessio Gasperetti
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
- University Heart Center, Zurich University Hospital, Zurich, Switzerland
| | - Ilaria Stadiotti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
14
|
Jackson EK, Gillespie DG, Tofovic SP. DPP4 Inhibition, NPY 1-36, PYY 1-36, SDF-1 α, and a Hypertensive Genetic Background Conspire to Augment Cell Proliferation and Collagen Production: Effects That Are Abolished by Low Concentrations of 2-Methoxyestradiol. J Pharmacol Exp Ther 2020; 373:135-148. [PMID: 32015161 PMCID: PMC7174788 DOI: 10.1124/jpet.119.263467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
By reducing their metabolism, dipeptidyl peptidase 4 inhibition (DPP4I) enhances the effects of numerous peptides including neuropeptide Y1–36 (NPY1–36), peptide YY1–36 (PYY1–36), and SDF-1α. Studies show that separately NPY1–36, PYY1–36 and SDF-1α stimulate proliferation of, and collagen production by, cardiac fibroblasts (CFs), preglomerular vascular smooth muscle cells (PGVSMCs), and glomerular mesangial cells (GMCs), particularly in cells isolated from genetically hypertensive rats. Whether certain combinations of these factors, in the absence or presence of DPP4I, are more profibrotic than others is unknown. Here we contrasted 24 different combinations of conditions (DPP4I, hypertensive genotype and physiologic levels [3 nM] of NPY1–36, PYY1–36, or SDF-1α) on proliferation of, and [3H]-proline incorporation by, CFs, PGVSMCs, and GMCs. In all three cell types, the various treatment conditions differentially increased proliferation and [3H]-proline incorporation, with a hypertensive genotype + DPP4I + NPY1–36 + SDF-1α being the most efficacious combination. Although the effects of this four-way combination were similar in male versus female CFs, physiologic (1 nM) concentrations of 2-methoxyestradiol (2ME; nonestrogenic metabolite of 17β-estradiol), abolished the effects of this combination in both male and female CFs. In conclusion, this study demonstrates that CFs, PGVSMCs, and GMCs are differentially activated by various combinations of NPY1–36, PYY1–36, SDF-1α, a hypertensive genetic background and DPP4I. We hypothesize that as these progrowth conditions accumulate, a tipping point would be reached that manifests in the long term as organ fibrosis and that 2ME would obviate any profibrotic effects of DPP4I, even under the most profibrotic conditions (i.e., hypertensive genotype with high NPY1–36 + SDF-1α levels and low 2ME levels).
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Delbert G Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stevan P Tofovic
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Medzikovic L, Aryan L, Eghbali M. Connecting sex differences, estrogen signaling, and microRNAs in cardiac fibrosis. J Mol Med (Berl) 2019; 97:1385-1398. [PMID: 31448389 DOI: 10.1007/s00109-019-01833-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/24/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Sex differences are evident in the pathophysiology of heart failure (HF). Progression of HF is promoted by cardiac fibrosis and no fibrosis-specific therapies are currently available. The fibrotic response is mediated by cardiac fibroblasts (CFs), and a central event is their phenotypic transition to pro-fibrotic myofibroblasts. These myofibroblasts may arise from various cellular origins including resident CFs and epicardial and endothelial cells. Both female subjects in clinical studies and female animals in experimental studies generally present less cardiac fibrosis compared with males. This difference is at least partially considered attributable to the ovarian hormone 17β-estradiol (E2). E2 signals via estrogen receptors to regulate genes are involved in the fibrotic response and myofibroblast transition. Besides protein-coding genes, E2 also regulates transcription of microRNA that modulate cardiac fibrosis. Sex dimorphism, E2, and miRNAs form multi-level regulatory networks in the pathophysiology of cardiac fibrosis, and the mechanism of these networks is not yet fully deciphered. Therefore, this review is aimed at summarizing current knowledge on sex differences, E2, and estrogen receptors in cardiac fibrosis, emphasizing on microRNAs and myofibroblast origins. KEY MESSAGES: • E2 and ERs regulate cardiac fibroblast function. • E2 and ERs may distinctly affect male and female cardiac fibrosis pathophysiology. • Sex, E2, and miRNAs form multi-level regulatory networks in cardiac fibrosis. • Sex-dimorphic and E2-regulated miRNAs affect mesenchymal transition.
Collapse
Affiliation(s)
- Lejla Medzikovic
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Laila Aryan
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
16
|
Weidner K, Behnes M, Rusnak J, Schupp T, Hoppner J, Taton G, Reiser L, Bollow A, Reichelt T, Ellguth D, Engelke N, Kuche P, Ansari U, El‐Battrawy I, Lang S, Nienaber CA, Akin M, Mashayekhi K, Ferdinand D, Weiß C, Borggrefe M, Akin I. Male sex increases mortality in ventricular tachyarrhythmias. Intern Med J 2019; 49:711-721. [DOI: 10.1111/imj.14170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Kathrin Weidner
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Michael Behnes
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Jonas Rusnak
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Tobias Schupp
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Jorge Hoppner
- Clinic for Diagnostic and Interventional Radiology HeidelbergUniversity of Heidelberg Heidelberg Germany
| | - Gabriel Taton
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Linda Reiser
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Armin Bollow
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Thomas Reichelt
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Dominik Ellguth
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Niko Engelke
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Philipp Kuche
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Uzair Ansari
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Ibrahim El‐Battrawy
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Siegfried Lang
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | | | - Muharrem Akin
- Department of Cardiology and AngiologyHannover Medical School Hannover Germany
| | - Kambis Mashayekhi
- Department of Cardiology and Angiology IIUniversity Heart Center Freiburg Bad Krozingen Germany
| | - Dennis Ferdinand
- Institute of Biomathematics and Medical Statistics, Faculty of Medicine MannheimUniversity Medical Center Mannheim Germany
| | | | - Martin Borggrefe
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Ibrahim Akin
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| |
Collapse
|
17
|
Groban L, Tran QK, Ferrario CM, Sun X, Cheng CP, Kitzman DW, Wang H, Lindsey SH. Female Heart Health: Is GPER the Missing Link? Front Endocrinol (Lausanne) 2019; 10:919. [PMID: 31993020 PMCID: PMC6970950 DOI: 10.3389/fendo.2019.00919] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
The G Protein-Coupled Estrogen Receptor (GPER) is a novel membrane-bound receptor that mediates non-genomic actions of the primary female sex hormone 17β-estradiol. Studies over the past two decades have elucidated the beneficial actions of this receptor in a number of cardiometabolic diseases. This review will focus specifically on the cardiac actions of GPER, since this receptor is expressed in cardiomyocytes as well as other cells within the heart and most likely contributes to estrogen-induced cardioprotection. Studies outlining the impact of GPER on diastolic function, mitochondrial function, left ventricular stiffness, calcium dynamics, cardiac inflammation, and aortic distensibility are discussed. In addition, recent data using genetic mouse models with global or cardiomyocyte-specific GPER gene deletion are highlighted. Since estrogen loss due to menopause in combination with chronological aging contributes to unique aspects of cardiac dysfunction in women, this receptor may provide novel therapeutic effects. While clinical studies are still required to fully understand the potential for pharmacological targeting of this receptor in postmenopausal women, this review will summarize the evidence gathered thus far on its likely beneficial effects.
Collapse
Affiliation(s)
- Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
- *Correspondence: Leanne Groban
| | - Quang-Kim Tran
- Department of Physiology & Pharmacology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, United States
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Che Ping Cheng
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Dalane W. Kitzman
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
18
|
Michalson KT, Groban L, Howard TD, Shively CA, Sophonsritsuk A, Appt SE, Cline JM, Clarkson TB, Carr JJ, Kitzman DW, Register TC. Estradiol Treatment Initiated Early After Ovariectomy Regulates Myocardial Gene Expression and Inhibits Diastolic Dysfunction in Female Cynomolgus Monkeys: Potential Roles for Calcium Homeostasis and Extracellular Matrix Remodeling. J Am Heart Assoc 2018; 7:e009769. [PMID: 30571375 PMCID: PMC6404177 DOI: 10.1161/jaha.118.009769] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
Background Left ventricular ( LV ) diastolic dysfunction often precedes heart failure with preserved ejection fraction, the dominant form of heart failure in postmenopausal women. The objective of this study was to determine the effect of oral estradiol treatment initiated early after ovariectomy on LV function and myocardial gene expression in female cynomolgus macaques. Methods and Results Monkeys were ovariectomized and randomized to receive placebo (control) or oral estradiol at a human-equivalent dose of 1 mg/day for 8 months. Monkeys then underwent conventional and tissue Doppler imaging to assess cardiac function, followed by transcriptomic and histomorphometric analyses of LV myocardium. Age, body weight, blood pressure, and heart rate were similar between groups. Echocardiographic mitral early and late inflow velocities, mitral annular velocities, and mitral E deceleration slope were higher in estradiol monkeys (all P<0.05), despite similar estimated LV filling pressure. MCP1 (monocyte chemoattractant protein 1) and LV collagen staining were lower in estradiol animals ( P<0.05). Microarray analysis revealed differential myocardial expression of 40 genes (>1.2-fold change; false discovery rate, P<0.05) in estradiol animals relative to controls, which implicated pathways associated with better calcium ion homeostasis and muscle contraction and lower extracellular matrix deposition ( P<0.05). Conclusions Estradiol treatment initiated soon after ovariectomy resulted in enhanced LV diastolic function, and altered myocardial gene expression towards decreased extracellular matrix deposition, improved myocardial contraction, and calcium homeostasis, suggesting that estradiol directly or indirectly modulates the myocardial transcriptome to preserve cardiovascular function.
Collapse
Affiliation(s)
- Kristofer T. Michalson
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Leanne Groban
- Department of AnesthesiologyWake Forest University School of MedicineWinston‐SalemNC
| | - Timothy D. Howard
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNC
| | - Carol A. Shively
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Areepan Sophonsritsuk
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Susan E. Appt
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - J. Mark Cline
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Thomas B. Clarkson
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - J. Jeffrey Carr
- Department of RadiologyVanderbilt University School of MedicineNashvilleTN
| | - Dalane W. Kitzman
- Section on CardiologyDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC
| | - Thomas C. Register
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| |
Collapse
|
19
|
do Carmo JM, da Silva AA, Moak SP, Browning JR, Dai X, Hall JE. Increased sleep time and reduced energy expenditure contribute to obesity after ovariectomy and a high fat diet. Life Sci 2018; 212:119-128. [PMID: 30273560 PMCID: PMC6240909 DOI: 10.1016/j.lfs.2018.09.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Abstract
In this study we examined if sleep time, caloric intake and energy expenditure are important contributors to development of ovariectomy-induced obesity in mice fed control or high fat diet (HFD). Twelve female mice at 6 weeks of age were divided into 2 groups: Sham (n = 5) and ovariectomized (OVX, n = 7). Mice were fed control diet for 9 weeks and shifted to HFD for additional 9 weeks. Food intake and body weight were measured daily and body composition was measured weekly by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), motor activity (MA) and sleep time were monitored at week 9 during control diet and HFD. OVX did not alter caloric intake, body weight or body composition, MA, sleep time or fasting blood glucose, but slightly reduced EE compared to Sham mice on control diet. After HFD feeding, OXV mice had similar caloric intake, lean mass, MA, and blood glucose levels but had significantly greater weight gain (8.2 ± 1.0 vs. 4.8 ± 1.2 g, p < 0.05), increased fat mass and sleep time, and reduced EE (3.3 ± 0.4 vs. 5.5 ± 0.2 kcal/h) and VO2 (1.12 ± 0.01 vs. 1.83 ± 0.05 ml/min) compared to Sham group. Daytime blood pressure was higher while nighttime heart rate was lower in OVX group. These results suggest that OVX may not substantially alter body weight or body composition in mice fed a normal diet, but when combined with HFD it increases sleep time and reduces EE, leading to greater weight gain and adiposity without altering food intake.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America.
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Sydney P Moak
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Jackson R Browning
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Xuemei Dai
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - John E Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States of America
| |
Collapse
|
20
|
Oza MJ, Kulkarni YA. Biochanin A improves insulin sensitivity and controls hyperglycemia in type 2 diabetes. Biomed Pharmacother 2018; 107:1119-1127. [PMID: 30257324 DOI: 10.1016/j.biopha.2018.08.073] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/14/2018] [Accepted: 08/15/2018] [Indexed: 01/25/2023] Open
Abstract
Biochanin A (5,7-Dihydroxy-4'-methoxyisoflavone) is an O-methylated isoflavone known for its anti-inflammatory, lipid lowering and anti-cancer activity. The current study was designed to find out antidiabetic efficacy of Biochanin A in type 2 diabetes in rats. Induction of type 2 diabetes mellitus in experimental animals was carried out by manipulation of diet using high fat diet for fourteen days and then administration of streptozotocin at low dose of 35 mg/kg, i.p. The diabetic animals were treated with 10, 20 and 40 mg/kg of Biochanin A for 28 days. The effect of Biochanin A treatment in diabetic animals was evaluated by measuring changes in body weight, biochemical parameters, insulin sensitivity index, Homeostatic model assessment-Insulin resistance (HOMA-IR), oral glucose tolerance test, glycohaemoglobin and hepatic glycogen level. Changes in histopathological characteristics of pancreatic tissue were also evaluated after treatment with Biochanin A. Immunohistochemical analysis of pancreatic tissue was carried out for the expression of SIRT1. The results showed that the selected doses of (10, 20 and 40 mg/kg) Biochanin A significantly decreased blood glucose (p < 0.001). The higher dose (40 mg/kg) of Biochanin A significantly reduced glucose tolerance (p < 0.001) in diabetic animals. Biochanin A treatment significantly reduced insulin resistance (p < 0.001) and improved inulin sensitivity (p < 0.01 for 10 mg/kg, 20 mg/kg, p < 0.001 for 40 mg/kg) at all selected dose levels. It also improved lipid profile significantly (p < 0.001) at lower, middle and higher dose level. Glycohaemoglobin formation was significantly decreased in diabetic animals (p < 0.001) after treatment with Biochanin A at all three dose levels. Liver glycogen level was also improved significantly after treatment with Biochanin A in diabetic animals at 20 mg/kg and 40 mg/kg dose level. Biochanin A at dose of 40 mg/kg increased SIRT1 expression in pancreatic tissue. In conclusion, Biochanin A has significant effect in type 2 diabetes mellitus which might be linked to its effects on SIRT1.
Collapse
Affiliation(s)
- Manisha J Oza
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India; SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
21
|
Maayah ZH, Levasseur J, Siva Piragasam R, Abdelhamid G, Dyck JRB, Fahlman RP, Siraki AG, El-Kadi AOS. 2-Methoxyestradiol protects against pressure overload-induced left ventricular hypertrophy. Sci Rep 2018; 8:2780. [PMID: 29426916 PMCID: PMC5807528 DOI: 10.1038/s41598-018-20613-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Numerous experimental studies have supported the evidence that 2-methoxyestradiol (2 ME) is a biologically active metabolite that mediates multiple effects on the cardiovascular system, largely independent of the estrogen receptor. 2 ME is a major cytochrome P450 1B1 (CYP1B1) metabolite and has been reported to have vasoprotective and anti-inflammatory actions. However, whether 2 ME would prevent cardiac hypertrophy induced by abdominal aortic constriction (AAC) has not been investigated yet. Therefore, the overall objectives of the present study were to elucidate the potential antihypertrophic effect of 2 ME and explore the mechanism(s) involved. Our results showed that 2 ME significantly inhibited AAC-induced left ventricular hypertrophy using echocardiography. The antihypertrophic effect of 2 ME was associated with a significant inhibition of CYP1B1 and mid-chain hydroxyeicosatetraenoic acids. Based on proteomics data, the protective effect of 2 ME is linked to the induction of antioxidant and anti-inflammatory proteins in addition to the modulation of proteins involved in myocardial energy metabolism. In vitro, 2 ME has shown a direct antihypertrophic effect through mitogen-activated protein kinases- and nuclear factor-κB-dependent mechanisms. The present work shows a strong evidence that 2 ME protects against left ventricular hypertrophy. Our data suggest the potential of repurposing 2 ME as a selective CYP1B1 inhibitor for the treatment of heart failure.
Collapse
Affiliation(s)
- Zaid H Maayah
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Jody Levasseur
- Cardiovascular Research Centre, Department of Pediatrics, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ramanaguru Siva Piragasam
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Ghada Abdelhamid
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Richard P Fahlman
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada.,Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Arno G Siraki
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada.
| |
Collapse
|
22
|
Sex Differences in Vascular Reactivity to Angiotensin II During the Evolution of Myocardial Infarction. J Cardiovasc Pharmacol 2018; 71:19-25. [DOI: 10.1097/fjc.0000000000000542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Iorga A, Cunningham CM, Moazeni S, Ruffenach G, Umar S, Eghbali M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ 2017; 8:33. [PMID: 29065927 PMCID: PMC5655818 DOI: 10.1186/s13293-017-0152-8] [Citation(s) in RCA: 464] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/04/2017] [Indexed: 12/15/2022] Open
Abstract
Epidemiologic studies have previously suggested that premenopausal females have reduced incidence of cardiovascular disease (CVD) when compared to age-matched males, and the incidence and severity of CVD increases postmenopause. The lower incidence of cardiovascular disease in women during reproductive age is attributed at least in part to estrogen (E2). E2 binds to the traditional E2 receptors (ERs), estrogen receptor alpha (ERα), and estrogen receptor beta (ERβ), as well as the more recently identified G-protein-coupled ER (GPR30), and can exert both genomic and non-genomic actions. This review summarizes the protective role of E2 and its receptors in the cardiovascular system and discusses its underlying mechanisms with an emphasis on oxidative stress, fibrosis, angiogenesis, and vascular function. This review also presents the sexual dimorphic role of ERs in modulating E2 action in cardiovascular disease. The controversies surrounding the clinical use of exogenous E2 as a therapeutic agent for cardiovascular disease in women due to the possible risks of thrombotic events, cancers, and arrhythmia are also discussed. Endogenous local E2 biosynthesis from the conversion of testosterone to E2 via aromatase enzyme offers a novel therapeutic paradigm. Targeting specific ERs in the cardiovascular system may result in novel and possibly safer therapeutic options for cardiovascular protection.
Collapse
Affiliation(s)
- Andrea Iorga
- Present address: Department of Medicine, Division of Gastroenterology/Liver, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
| | - Christine M Cunningham
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Shayan Moazeni
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Gregoire Ruffenach
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
24
|
Okazaki M, Kaneko M, Ishida Y, Murase N, Katsumura T. Changes in the Width of the Tibiofibular Syndesmosis Related to Lower Extremity Joint Dynamics and Neuromuscular Coordination on Drop Landing During the Menstrual Cycle. Orthop J Sports Med 2017; 5:2325967117724753. [PMID: 28913369 PMCID: PMC5590700 DOI: 10.1177/2325967117724753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Many injuries of the lower extremities, especially the knee and ankle, occur during sports activity, and the incidence rate is higher in women than in men. Hypothesis: The hypothesis was that phases of the menstrual cycle affect the width of the tibiofibular syndesmosis during drop landing in healthy young women and that such changes at the tibiofibular joint also affect the dynamics and neuromuscular coordination of the lower extremities. Study Design: Descriptive laboratory study. Methods: Participants included 28 healthy young women (mean age, 21.0 ± 0.8 years). Blood samples were collected to determine plasma levels of estradiol and progesterone immediately before the performance of the task: drop landing on a single leg from a 30-cm platform. Using ultrasonography, the distance between the tibia and the distal end of the fibula, regarded as the width of the tibiofibular syndesmosis, was measured in an upright position without flexion of the ankle. The peak ground-reaction force (GRF) on landing was measured using a force platform. The time to peak GRF (Tp-GRF) was measured as the time from initial ground contact to the peak GRF. Hip, knee, and ankle joint angles during the single-leg landing were calculated using a 3-dimensional motion analysis system. Muscle activities of the lower extremities were measured using surface electromyography. Results: The width of the tibiofibular syndesmosis was significantly greater in the luteal phase when compared with the menstrual, follicular, and ovulation phases (by 5%-8% of control). Also, during the luteal phase, the Tp-GRF was significantly shorter than in the follicular phase (by 6%); hip internal rotation and knee valgus were significantly greater than in the menstrual phase (by 43% and 34%, respectively); knee flexion was significantly less than in the menstrual and follicular phases (by 7%-9%); ankle dorsiflection was significantly less than in the follicular phase (by 11%); ankle adduction and eversion were significantly greater than in the menstrual and follicular phases (by 26%-46%, and 27%-33%, respectively); and activation of the gluteus maximus before landing was significantly lower than in the menstrual and follicular phases (by 20%-22%). Conclusion: The luteal phase appears to be associated with decreased strength and laxity of the ankle as well as lower extremity muscle activity in women. The changes presumably represent a greater risk for sports injuries. Clinical Relevance: The results of this study suggest that the luteal phase may be related to the greater incidence of lower extremity injuries in women.
Collapse
Affiliation(s)
| | | | - Yukisato Ishida
- Department of Physical Therapy, Bunkyo Gakuin University, Tokyo, Japan
| | - Norio Murase
- Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan
| | | |
Collapse
|
25
|
InanlooRahatloo K, Liang G, Vo D, Ebert A, Nguyen I, Nguyen PK. Sex-based differences in myocardial gene expression in recently deceased organ donors with no prior cardiovascular disease. PLoS One 2017; 12:e0183874. [PMID: 28850583 PMCID: PMC5574577 DOI: 10.1371/journal.pone.0183874] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/12/2017] [Indexed: 02/08/2023] Open
Abstract
Sex differences in the development of the normal heart and the prevalence of cardiomyopathies have been reported. The molecular basis of these differences remains unclear. Sex differences in the human heart might be related to patterns of gene expression. Recent studies have shown that sex specific differences in gene expression in tissues including the brain, kidney, skeletal muscle, and liver. Similar data is limited for the heart. Herein we address this issue by analyzing donor and post-mortem adult human heart samples originating from 46 control individuals to study whole-genome gene expression in the human left ventricle. Using data from the genotype tissue expression (GTEx) project, we compared the transcriptome expression profiles of male and female hearts. We found that genes located on sex chromosomes were the most abundant ones among the sexually dimorphic genes. The majority of differentially expressed autosomal genes were those involved in the regulation of inflammation, which has been found to be an important contributor to left ventricular remodeling. Specifically, genes on autosomal chromosomes encoding chemokines with inflammatory functions (e.g. CCL4, CX3CL1, TNFAIP3) and a gene that regulates adhesion of immune cells to the endothelium (e.g., VCAM1) were identified with sex-specific expression levels. This study underlines the relevance of sex as an important modifier of cardiac gene expression. These results have important implications in the understanding of the differences in the physiology of the male and female heart transcriptome and how they may lead to different sex specific difference in human cardiac health and its control.
Collapse
Affiliation(s)
- Kolsoum InanlooRahatloo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Genetics, Genetic Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Grace Liang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Davis Vo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Antje Ebert
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ivy Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States of America
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University and Veterans Affairs Palo Alto, Palo Alto, California, United States of America
- * E-mail: ,
| |
Collapse
|
26
|
Nio AQX, Stöhr EJ, Shave RE. Age-related differences in left ventricular structure and function between healthy men and women. Climacteric 2017; 20:476-483. [DOI: 10.1080/13697137.2017.1356814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- A. Q. X. Nio
- Department of Physiology and Health, Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, UK
| | - E. J. Stöhr
- Department of Physiology and Health, Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, UK
| | - R. E. Shave
- Department of Physiology and Health, Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, UK
| |
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
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 417] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
29
|
Schwertz DW, Beck JM, Kowalski JM, Ross JD. Sex Differences in the Response of Rat Heart Ventricle to Calcium. Biol Res Nurs 2016; 5:286-98. [PMID: 15068658 DOI: 10.1177/1099800403262615] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Calcium (Ca2+ ) is a key mediator of myocardial function. Calcium regulates contraction, and disruption of myocellular Ca2+ handling plays a role in cardiac pathologies such as arrhythmias and heart failure. This investigation examines sex differences in sensitivity of the contractile proteins to Ca2+ and myofibrillar Ca2+ delivery in the ventricular myocardium. Sensitivity of contractile proteins to Ca2+ was measured in weight-matched male and female Sprague-Dawley rats using the skinned ventricular papillary muscle fiber and Ca2+ -stimulated Mg2+ -dependent adenosine triphosphatase (ATPase) activity methodologies. Calcium delivery was examined by measuring the contractile response to a range of extracellular Ca2+ concentrations in isolated ventricular myocytes, papillary muscle, and the isolated perfused whole heart. Findings from studies in the whole heart suggest that at a fixed preload, the male left ventricle generates more pressure than a female ventricle over a range of extracellular Ca2+ concentrations. In contrast, results from myocyte and papillary muscle studies suggest that females require less extracellular Ca2+ to elicit a similar contractile response. Results obtained from the 2 methods used to determine sex differences in Ca2+ sensitivity were equivocal. Further studies are required to elucidate sex differences in myocardial Ca2+ handling and the reasons for disparate results in different heart muscle preparations. The results of these studies will lead to the design of sex-optimized therapeutic interventions for cardiac disease.
Collapse
Affiliation(s)
- Dorie W Schwertz
- College of Nursing M/C 802, University of Illinois, at Chicago, 845 South Damen Avenue, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
30
|
Olszanecka A, Kawecka-Jaszcz K, Czarnecka D. Association of free testosterone and sex hormone binding globulin with metabolic syndrome and subclinical atherosclerosis but not blood pressure in hypertensive perimenopausal women. Arch Med Sci 2016; 12:521-8. [PMID: 27279843 PMCID: PMC4889686 DOI: 10.5114/aoms.2016.59925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/20/2015] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Data on the role of androgens as potential mediators of increasing cardiovascular risk in women at midlife are controversial. The aim of the study was to analyze the relationship of free testosterone (FT) and sex hormone binding globulin (SHBG) with blood pressure and subclinical organ damage and metabolic syndrome (MS) in middle aged hypertensive women. MATERIAL AND METHODS One hundred and fifty-two women with newly diagnosed arterial hypertension were included in the study. In all subjects blood pressure measurements were performed as well as echocardiographic examination with left ventricular structure and function assessment (GE Vivid 7.0), carotid ultrasound with measurement of intima-media thickness (IMT), and carotid-femoral pulse wave velocity (PWV) measurement (Sphygmocor). A fasting blood sample was taken to measure glucose and lipid concentrations. Serum testosterone and SHBG were measured. Free testosterone was calculated according to the Vermeulen formula. Metabolic syndrome was defined following the International Diabetes Federation (IDF) recommendations. RESULTS Free testosterone was significantly higher and SHBG lower in women with MS independently of menopausal status. The odds ratio of MS per quartile increment in FT after adjustment for covariates was 2.06 (95% CI: 1.16-3.65). There was no correlation between FT, SHBG and blood pressure. Free testosterone was associated with decreased left ventricular diastolic function (E/A ratio β = -0.19, p = 0.05) and subclinical atherosclerosis (IMT β = 0.34, p = 0.009), but not arterial stiffness. CONCLUSIONS Free testosterone and SHBG independently of menopause status are related to MS. Free testosterone is associated with worse metabolic profile, subclinical atherosclerosis and impaired diastolic function of the left ventricle.
Collapse
Affiliation(s)
- Agnieszka Olszanecka
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Kalina Kawecka-Jaszcz
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Danuta Czarnecka
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
31
|
Huang CK, Lee SO, Chang E, Pang H, Chang C. Androgen receptor (AR) in cardiovascular diseases. J Endocrinol 2016; 229:R1-R16. [PMID: 26769913 PMCID: PMC4932893 DOI: 10.1530/joe-15-0518] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/13/2016] [Indexed: 01/13/2023]
Abstract
Cardiovascular diseases (CVDs) are still the highest leading cause of death worldwide. Several risk factors have been linked to CVDs, including smoking, diabetes, hyperlipidemia, and gender among others. Sex hormones, especially the androgen and its receptor, androgen receptor (AR), have been linked to many diseases with a clear gender difference. Here, we summarize the effects of androgen/AR on CVDs, including hypertension, stroke, atherosclerosis, abdominal aortic aneurysm (AAA), myocardial hypertrophy, and heart failure, as well as the metabolic syndrome/diabetes and their impacts on CVDs. Androgen/AR signaling exacerbates hypertension, and anti-androgens may suppress hypertension. Androgen/AR signaling plays dual roles in strokes, depending on different kinds of factors; however, generally males have a higher incidence of strokes than females. Androgen and AR differentially modulate atherosclerosis. Androgen deficiency causes elevated lipid accumulation to enhance atherosclerosis; however, targeting AR in selective cells without altering serum androgen levels would suppress atherosclerosis progression. Androgen/AR signaling is crucial in AAA development and progression, and targeting androgen/AR profoundly restricts AAA progression. Men have increased cardiac hypertrophy compared with age-matched women that may be due to androgens. Finally, androgen/AR plays important roles in contributing to obesity and insulin/leptin resistance to increase the metabolic syndrome.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- George Whipple Lab for Cancer ResearchDepartments of Pathology, Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Soo Ok Lee
- George Whipple Lab for Cancer ResearchDepartments of Pathology, Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Eugene Chang
- George Whipple Lab for Cancer ResearchDepartments of Pathology, Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA Department of MedicineCase Cardiovascular Institute Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Haiyan Pang
- George Whipple Lab for Cancer ResearchDepartments of Pathology, Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer ResearchDepartments of Pathology, Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA Sex Hormone Research CenterChina Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
32
|
Antonio EL, Serra AJ, dos Santos AA, Vieira SS, Silva JMA, Yoshizaki A, Sofia RR, Tucci PJF. Are there gender differences in left ventricular remodeling after myocardial infarction in rats? REVISTA BRASILEIRA DE CIRURGIA CARDIOVASCULAR : ORGAO OFICIAL DA SOCIEDADE BRASILEIRA DE CIRURGIA CARDIOVASCULAR 2015; 30:70-6. [PMID: 25859870 PMCID: PMC4389530 DOI: 10.5935/1678-741.20140093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 06/02/2014] [Indexed: 11/21/2022]
Abstract
Objective An unclear issue is whether gender may influence at cardiac remodeling after
myocardial infarction (MI). We evaluated left ventricle remodeling in female
and male rats post-MI. Methods Rats were submitted to anterior descending coronary occlusion.
Echocardiographic evaluations were performed on the first and sixth week
post-occlusion to determine myocardial infarction size and left ventricle
systolic function (FAC, fractional area change). Pulsed Doppler was applied
to analyze left ventricle diastolic function using the following parameters:
E wave, A wave, E/A ratio. Two-way ANOVA was applied for comparisons,
complemented by the Bonferroni test. A P≤=0.05 was
considered significant. Results There were no significant differences between genders for morphometric
parameters on first (MI [Female (FE): 44.0±5.0 vs. Male (MA): 42.0±3.0%];
diastolic [FE: 0.04±0.003 vs. MA: 0.037±0.005, mm/g] and systolic [FE:
0.03±0.0004 vs. MA: 0.028±0.005, mm/g] diameters of left ventricle) and
sixth (MI [FE: 44.0±5.0 vs. MA: 42.0±3.0, %]; diastolic [FE: 0.043±0.01 vs.
MA: 0.034±0.005, mm/g] and systolic [FE: 0.035±0.01 vs. MA: 0.027±0.005,
mm/g] of LV) week. Similar findings were reported for left ventricle
functional parameters on first (FAC [FE: 34.0±6.0 vs. MA: 32.0±4.0, %]; wave
E [FE: 70.0±18.0 vs. MA: 73.0±14.0, cm/s]; wave A [FE: 20.0±12.0 vs. MA:
28.0±13.0, cm/s]; E/A [FE: 4.9±3.4 vs. MA: 3.3±1.8]) and sixth (FAC [FE:
29.0±7.0 vs. MA: 31.0±7.0, %]; wave E [FE: 85.0±18.0 vs. MA: 87.0±20.0,
cm/s]; wave A [FE: 20.0±11.0 vs. MA: 28.0±17.0, cm/s]; E/A [FE: 6.2±4.0 vs.
MA: 4.6±3.4]) week. Conclusion Gender does not influence left ventricle remodeling post-MI in rats.
Collapse
Affiliation(s)
- Ednei Luiz Antonio
- Laboratório de Fisiologia e Fisiopatologia Cardíacas, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | - Stella Sousa Vieira
- Laboratório de Fisiologia e Fisiopatologia Cardíacas, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Amanda Yoshizaki
- Programa de Pós-graduação em Ciências da Reabilitação, Universidade Nove de Julho, São Paulo, SP, Brazil
| | | | - Paulo José Ferreira Tucci
- Laboratório de Fisiologia e Fisiopatologia Cardíacas, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Salerni S, Di Francescomarino S, Cadeddu C, Acquistapace F, Maffei S, Gallina S. The different role of sex hormones on female cardiovascular physiology and function: not only oestrogens. Eur J Clin Invest 2015; 45:634-45. [PMID: 25845675 DOI: 10.1111/eci.12447] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/31/2015] [Indexed: 12/18/2022]
Abstract
Human response to different physiologic stimuli and cardiovascular (CV) adaptation to various pathologies seem to be gender specific. Sex-steroid hormones have been postulated as the major contributors towards these sex-related differences. This review will discuss current evidence on gender differences in CV function and remodelling, and will present the different role of the principal sex-steroid hormones on female heart. Starting from a review of sex hormones synthesis, receptors and CV signalling, we will summarize the current knowledge concerning the role of sex hormones on the regulation of our daily activities throughout the life, via the modulation of autonomic nervous system, excitation-contraction coupling pathway and ion channels activity. Many unresolved questions remain even if oestrogen effects on myocardial remodelling and function have been extensively studied. So this work will focus attention also on the controversial and complex relationship existing between androgens, progesterone and female heart.
Collapse
Affiliation(s)
- Sara Salerni
- Department of Neuroimaging, University of Chieti, Chieti, Italy
| | | | - Christian Cadeddu
- Department of Cardiovascular and Neurological Sciences, University of Cagliari, Cagliari, Italy
| | - Flavio Acquistapace
- Helvetic Confederation IIHM International institute, Cardiologic Medical Center Manno, Lugano, Helvetia
| | - Silvia Maffei
- Fondazione G. Monasterio CNR-Regione Toscana and Institute of Clinical Physiology-CNR, Pisa, Italy
| | - Sabina Gallina
- Department of Neuroimaging, University of Chieti, Chieti, Italy
| |
Collapse
|
34
|
Wang H, Zhao Z, Lin M, Groban L. Activation of GPR30 inhibits cardiac fibroblast proliferation. Mol Cell Biochem 2015; 405:135-48. [PMID: 25893735 DOI: 10.1007/s11010-015-2405-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/09/2015] [Indexed: 12/28/2022]
Abstract
The incidence of left ventricular diastolic dysfunction significantly increases in postmenopausal women suggesting the association between estrogen loss and diastolic dysfunction. The in vivo activation of G protein-coupled estrogen receptor (GPR30) attenuates the adverse effects of estrogen loss on cardiac fibrosis and diastolic dysfunction in mRen2.Lewis rats. This study was designed to address the effects of GPR30 on cardiac fibroblast proliferation in rats. The expression of GPR30 in cardiac fibroblasts isolated from adult Sprague-Dawley rats was confirmed by RT-PCR, Western blot analysis, and immunofluorescence staining. Results from BrdU incorporation assays, cell counting, carboxyfluorescein diacetate succinimidyl ester labeling in conjunction with flow cytometry, and Ki-67 staining showed that treatment with G1, a specific agonist of GPR30, inhibited cardiac fibroblast proliferation in a dose-dependent manner, which was associated with decreases in CDK1 and cyclin B1 protein expressions. In the GPR30-KO cells, BrdU incorporation, and CDK1 and cyclin B1 expressions significantly increased when compared to GPR30-intact cells. G1 had no effect on BrdU incorporation, CDK1 and cyclin B1 mRNA levels in GPR30-KO cells. In vivo studies showed increases in CDK1 and cyclin B1 mRNA levels, Ki-67-positive cells, and the immunohistochemistry staining of vimentin, a fibroblast marker, in the left ventricles from ovariectomized mRen2.Lewis rats versus hearts from ovary-intact littermates; 2 weeks of G1 treatment attenuated these adverse effects of estrogen loss. This study demonstrates that GPR30 is expressed in rat cardiac fibroblasts, and activation of GPR30 limits proliferation of these cells likely via suppression of the cell cycle proteins, cyclin B1, and CDK1.
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1009, USA
| | | | | | | |
Collapse
|
35
|
Merino PM, Martínez D, Iñiguez G, Lopez P, Cassorla F, Perez-Bravo F, Codner E. Elevation of C-reactive protein during the luteal phase in healthy adolescents. Gynecol Endocrinol 2015; 31:260-3. [PMID: 25392126 DOI: 10.3109/09513590.2014.982086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Variations in inflammatory markers have been reported in adult women during the luteal phase, but whether these findings are observed during adolescence is unknown. We postulate that higher ultrasensitive C-reactive protein (usCRP) and lower 2-hydroxyestrone (2OHE) levels, an estrogen metabolite with cardioprotective actions, are present during the luteal phase in young women. AIM To evaluate usCRP levels during the menstrual cycle and to determine its association with 2OHE and 16α-hydroxyestrone (16OHE) in adolescents. METHODS Healthy postmenarcheal adolescents (N = 37) were studied during one menstrual cycle in follicular phase (FP) and luteal phase-like period (LP-L). RESULTS Elevations in usCRP levels in the LP-L were observed in the entire group and in anovulatory cycles (1.9 ± 1.1 mg/L in FP to 2.5 ± 1.8 mg/L in LP-L; p < 0.0001). Increases in estrone, estradiol, free and bioavailable estradiol, testosterone, usCRP and 2OHE levels were observed in LP-L compared with FP (p < 0.01), with a borderline elevation in IFG-I levels (p = 0.06). CONCLUSIONS We report an elevation of usCRP and 2OHE levels during the luteal phase in healthy adolescents. Elevations of this inflammatory marker in anovulatory adolescents without an increase in 2OHE may play a role in metabolic risks associated with chronic anovulation.
Collapse
Affiliation(s)
- Paulina M Merino
- Faculty of Medicine, Institute of Maternal and Child Research, University of Chile , Santiago , Chile
| | | | | | | | | | | | | |
Collapse
|
36
|
Li J, Jubair S, Janicki JS. Estrogen inhibits mast cell chymase release to prevent pressure overload-induced adverse cardiac remodeling. Hypertension 2014; 65:328-34. [PMID: 25403608 DOI: 10.1161/hypertensionaha.114.04238] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Estrogen regulation of myocardial chymase and chymase effects on cardiac remodeling are unknown. To test the hypothesis that estrogen prevents pressure overload-induced adverse cardiac remodeling by inhibiting mast cell (MC) chymase release, transverse aortic constriction or sham surgery was performed in 7-week-old intact and ovariectomized (OVX) rats. Three days before creating the constriction, additional groups of OVX rats began receiving 17β-estradiol, a chymase inhibitor, or a MC stabilizer. Left ventricular function, cardiomyocyte size, collagen volume fraction, MC density and degranulation, and myocardial and plasma chymase levels were assessed 18 days postsurgery. Aortic constriction resulted in ventricular hypertrophy in intact and OVX groups, whereas collagen volume fraction was increased only in OVX rats. Chymase protein content was increased by aortic constriction in the intact and OVX groups, with the magnitude of the increase being greater in OVX rats. MC density and degranulation, plasma chymase levels, and myocardial active transforming growth factor-β1 levels were increased by aortic constriction only in OVX rats. Estrogen replacement markedly attenuated the constriction-increased myocardial chymase, MC density and degranulation, plasma chymase, and myocardial active transforming growth factor-β1, as well as prevented ventricular hypertrophy and increased collagen volume fraction. Chymostatin attenuated the aortic constriction-induced ventricular hypertrophy and collagen volume fraction in the OVX rats similar to that achieved by estrogen replacement. Nedocromil yielded similar effects, except for the reduction of chymase content. We conclude that the estrogen-inhibited release of MC chymase is responsible for the cardioprotection against transverse aortic constriction-induced adverse cardiac remodeling.
Collapse
Affiliation(s)
- Jianping Li
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia
| | - Shaiban Jubair
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia
| | - Joseph S Janicki
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia.
| |
Collapse
|
37
|
Lee TM, Lin SZ, Chang NC. Both GPER and membrane oestrogen receptor-α activation protect ventricular remodelling in 17β oestradiol-treated ovariectomized infarcted rats. J Cell Mol Med 2014; 18:2454-65. [PMID: 25256868 PMCID: PMC4302651 DOI: 10.1111/jcmm.12430] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 08/19/2014] [Indexed: 12/20/2022] Open
Abstract
Clinical and experimental studies have established that gender is a factor in the development of ventricular hypertrophy. We investigated whether the attenuated hypertrophic effect of oestradiol was via activation of phosphatidylinositol 3-kinase (PI3K)/Akt/endothelial nitric oxide synthase (eNOS) through non-genomic action. Twenty-four hours after coronary ligation, female Wistar rats were randomized into control, subcutaneous oestradiol treatment or a G-protein coupled oestrogen receptor (GPER) agonist, G-1 and treated for 4 weeks starting from 2 weeks after bilateral ovariectomy. Ventricular hypertrophy assessed by cardiomyocyte size after infarction was similarly attenuated by oestradiol or G-1 in infarcted rats. The phosphorylation of Akt and eNOS was significantly decreased in infarcted rats and restored by oestradiol and G-1, implying the GPER pathway in this process. Oestradiol-induced Akt phosphorylation was not abrogated by G-15 (a GPER blocker). Akt activation was not inhibited by actinomycin D. When a membrane-impermeable oestrogen-albumin construct was applied, similar responses in terms of eNOS activation to those of oestradiol were achieved. Furthermore, PPT, an ERα receptor agonist, activated the phosphorylation of Akt and eNOS. Thus, membrane ERα receptor played a role in mediating the phosphorylation of Akt and eNOS. The specific PI3K inhibitor, LY290042, completely abolished Akt activation and eNOS phosphorylation in infarcted hearts treated with either oestradiol or oestradiol + G-15. These data support the conclusions that oestradiol improves ventricular remodelling by both GPER- and membrane-bound ERα-dependent mechanisms that converge into the PI3K/Akt/eNOS pathway, unveiling a novel mechanism by which oestradiol regulates pathological cardiomyocyte growth after infarction.
Collapse
Affiliation(s)
- Tsung-Ming Lee
- Department of Medicine, Cardiology Section, China Medical University-An Nan Hospital, Tainan, Taiwan; Department of Medicine, China Medical University, Taichung, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | |
Collapse
|
38
|
Lušin TT, Mrhar A, Marc J, Trontelj J, Zavratnik A, Zegura B, Pfeifer M, Ostanek B. Inverse correlation of carotid intima-media thickness with raloxifene serum levels in osteoporosis. Wien Klin Wochenschr 2014; 126:403-8. [PMID: 24842749 DOI: 10.1007/s00508-014-0551-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/13/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Raloxifene is a selective oestrogen receptor modulator with effects on bone and breast cancer and cardiovascular disease risk. The aim of this study was to examine the influence of raloxifene treatment on surrogate markers of atherosclerosis and the correlation of these markers with raloxifene serum concentrations. METHODS A prospective clinical trial on 53 postmenopausal osteoporotic women treated with raloxifene was performed. Surrogate markers of atherosclerosis (flow-mediated vasodilatation, glyceryltrinitrate-induced vasodilatation of the brachial artery, carotid intima-media thickness (c-IMT), inter-cell adhesion molecule-1, vascular-cell adhesion molecule-1 and E-selectin) were measured before and after 6 months of treatment. Serum concentrations of raloxifene and raloxifene metabolites were assessed after 12 months of treatment. The tested markers were correlated with measured serum concentrations of raloxifene species. RESULTS Among the tested surrogate markers of atherosclerosis c-IMT, E-selectin and ICAM changed significantly during treatment. A negative correlation of the non-metabolized raloxifene serum levels with the percentage change of c-IMT during treatment (r = - 0.315, p = 0.048) was found. Likewise, the sum of the levels of three raloxifene metabolites, raloxifene-6-b-glucuronide (M1), raloxifene-4'-b-glucuronide (M2) and raloxifene-6,4'-diglucuronide (M3) in serum showed a negative correlation with the percentage change of c-IMT during treatment (r = - 0.375, p = 0.017). For the other tested parameters, no correlation with raloxifene serum levels was found. CONCLUSIONS To the best of our knowledge, this is the first study correlating raloxifene species serum concentrations with changes in the surrogate markers of atherosclerosis. A greater decrease of c-IMT in patients with higher raloxifene concentrations could contribute to a lower risk of cardiovascular events in these patients.
Collapse
Affiliation(s)
- Tina Trdan Lušin
- Department of Biopharmacy and Pharmacokinetics, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Yorifuji T, Uchida T, Abe H, Toyofuku Y, Tamaki M, Fujitani Y, Hirose T, Kawamori R, Takeda S, Watada H. 2-Methoxyestradiol ameliorates glucose tolerance with the increase in β-cell mass in db/db mice. J Diabetes Investig 2014; 2:180-5. [PMID: 24843481 PMCID: PMC4014916 DOI: 10.1111/j.2040-1124.2010.00087.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aims/Introduction: 2‐Methoxyestradiol (2ME) is an estradiol metabolite with little estrogenic activity. Previous data identified its anti‐carcinogenic properties and possible cardiovascular benefits. However, its effect on diabetes mellitus has not been fully elucidated. The aim of the present study was to determine the effects of 2ME on glucose metabolism in the diabetic state. Materials and Methods: To evaluate the effects of 2ME, pellets of two different doses of the drug were implanted into female db/db mice at the age of 5 weeks. Intraperitoneal glucose tolerance test and insulin tolerance test were carried out at the age of 8 weeks. The pancreas was harvested for morphological analysis and β‐cell function at the age of 9 weeks. Results: 2ME improved random blood glucose levels and glucose tolerance with increases in insulin levels during an intraperitoneal glucose tolerance test. Insulin sensitivity judged by an insulin tolerance test was comparable in the low‐ and high‐dose 2ME groups and the control group. Although glucose‐stimulated insulin secretion in isolated islets was comparable among the three groups, β‐cell mass in 2ME‐treated groups was higher than the control group. In the 2ME‐treated groups, the number of Ki67‐positive cells in islets was higher, whereas the number of cleaved caspase‐3‐positive cells was comparable with the control. Conclusions: 2ME ameliorates glucose tolerance by promoting the proliferation of β‐cell mass in db/db mice. Our data suggests its potential clinical usefulness as a disease‐modifying drug for type 2 diabetes mellitus. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00087.x, 2011)
Collapse
Affiliation(s)
| | | | | | | | | | - Yoshio Fujitani
- Medicine, Metabolism and Endocrinology ; Center for Therapeutic Innovations in Diabetes
| | - Takahisa Hirose
- Medicine, Metabolism and Endocrinology ; Center for Therapeutic Innovations in Diabetes
| | - Ryuzo Kawamori
- Medicine, Metabolism and Endocrinology ; Center for Therapeutic Innovations in Diabetes ; Center for Beta Cell Biology and Regeneration
| | | | - Hirotaka Watada
- Medicine, Metabolism and Endocrinology ; Center for Beta Cell Biology and Regeneration
| |
Collapse
|
40
|
House M, Tadesse-Telila S, Norwitz ER, Socrate S, Kaplan DL. Inhibitory effect of progesterone on cervical tissue formation in a three-dimensional culture system with human cervical fibroblasts. Biol Reprod 2014; 90:18. [PMID: 24285720 DOI: 10.1095/biolreprod.113.112540] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Progesterone supplementation is recommended to prevent preterm birth in women with a short cervix, but the mechanism is unclear. We hypothesize that progesterone acts by altering the composition of the cervical extracellular matrix (ECM). We tested this hypothesis using human cervical fibroblasts in both two-dimensional (2D) and three-dimensional (3D) cultures. For 2D culture, cells were seeded in 6-well plates and cultured with media supplemented with estradiol (10(-8) M), progesterone (10(-7) or 10(-6) M), and vehicle. For 3D culture, the cells were cultured on a porous silk protein scaffold system. Progesterone and estrogen receptors were documented by immunohistochemistry and Western blot analysis. In both 2D and 3D cultures, decreased collagen synthesis was seen with increased progesterone concentration. Three-dimensional cultures could be maintained significantly longer than 2D cultures, and the morphology of 3D cultures appeared similar to native cervical tissue. Thus, further studies were performed in 3D culture. To determine the effect of progesterone concentration, the 3D scaffolds were cultured with estradiol (10(-8) M) and five conditions: vehicle; 10(-9), 10(-8), or 10(-7) M progesterone; or 10(-7) M progesterone plus 10(-6) M mifepristone. The highest progesterone concentration correlated with the least amount of collagen synthesis. Collagen synthesis progressively increased as progesterone concentration decreased. This effect was partially antagonized by mifepristone, suggesting the mechanism is mediated by the progesterone receptor. This hormonally responsive 3D culture system supports the hypothesis that progesterone has a direct effect on remodeling cervical ECM during pregnancy. The 3D culture system could be useful for studying the mechanism of progesterone effects on the cervix.
Collapse
Affiliation(s)
- Michael House
- Division of Maternal Fetal Medicine, Tufts Medical Center, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
41
|
Janicki JS, Spinale FG, Levick SP. Gender differences in non-ischemic myocardial remodeling: are they due to estrogen modulation of cardiac mast cells and/or membrane type 1 matrix metalloproteinase. Pflugers Arch 2013; 465:687-97. [PMID: 23417570 DOI: 10.1007/s00424-013-1229-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/14/2013] [Accepted: 01/28/2013] [Indexed: 10/27/2022]
Abstract
This review is focused on gender differences in cardiac remodeling secondary to sustained increases in cardiac volume (VO) and generated pressure (PO). Estrogen has been shown to favorably alter the course of VO-induced remodeling. That is, the VO-induced increased extracellular matrix proteolytic activity and mast cell degranulation responsible for the adverse cardiac remodeling in males and ovariectomized rodents do not occur in intact premenopausal females. While less is known regarding the mechanisms responsible for female cardioprotection in PO-induced stress, gender differences in remodeling have been reported indicating the ability of premenopausal females to adequately compensate. In view of the fact that, in male mice with PO, mast cells have been shown to play a role in the adverse remodeling suggests favorable estrogen modification of mast cell phenotype may also be responsible for cardioprotection in females with PO. Thus, while evidence is accumulating regarding premenopausal females being cardioprotected, there remains the need for in-depth studies to identify critical downstream molecular targets that are under the regulation of estrogen and relevant to cardiac remodeling. Such studies would result in the development of therapy which provides cardioprotection while avoiding the adverse effects of systemic estrogen delivery.
Collapse
Affiliation(s)
- Joseph S Janicki
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| | | | | |
Collapse
|
42
|
Regitz-Zagrosek V, Dworatzek E, Kintscher U, Dragun D. Sex and Sex Hormone–Dependent Cardiovascular Stress Responses. Hypertension 2013. [DOI: 10.1161/hypertensionaha.111.189233] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vera Regitz-Zagrosek
- From the Institute of Gender in Medicine (V.R.-Z., E.D.), Center for Cardiovascular Research (V.R.-Z., E.D., U.K., D.D.), Department of Pharmacology (U.K.), and Clinic for Nephrology and Intensive Care Medicine (D.D.), Charité Universitaetsmedizin, Berlin, Germany
| | - Elke Dworatzek
- From the Institute of Gender in Medicine (V.R.-Z., E.D.), Center for Cardiovascular Research (V.R.-Z., E.D., U.K., D.D.), Department of Pharmacology (U.K.), and Clinic for Nephrology and Intensive Care Medicine (D.D.), Charité Universitaetsmedizin, Berlin, Germany
| | - Ulrich Kintscher
- From the Institute of Gender in Medicine (V.R.-Z., E.D.), Center for Cardiovascular Research (V.R.-Z., E.D., U.K., D.D.), Department of Pharmacology (U.K.), and Clinic for Nephrology and Intensive Care Medicine (D.D.), Charité Universitaetsmedizin, Berlin, Germany
| | - Duska Dragun
- From the Institute of Gender in Medicine (V.R.-Z., E.D.), Center for Cardiovascular Research (V.R.-Z., E.D., U.K., D.D.), Department of Pharmacology (U.K.), and Clinic for Nephrology and Intensive Care Medicine (D.D.), Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
43
|
Distinct cardiac transcriptional profiles defining pregnancy and exercise. PLoS One 2012; 7:e42297. [PMID: 22860109 PMCID: PMC3409173 DOI: 10.1371/journal.pone.0042297] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/02/2012] [Indexed: 12/11/2022] Open
Abstract
Background Although the hypertrophic responses of the heart to pregnancy and exercise are both considered to be physiological processes, they occur in quite different hormonal and temporal settings. In this study, we have compared the global transcriptional profiles of left ventricular tissues at various time points during the progression of hypertrophy in exercise and pregnancy. Methodology/Principal Findings The following groups of female mice were analyzed: non-pregnant diestrus cycle sedentary control, mid-pregnant, late-pregnant, and immediate-postpartum, and animals subjected to 7 and 21 days of voluntary wheel running. Hierarchical clustering analysis shows that while mid-pregnancy and both exercise groups share the closest relationship and similar gene ontology categories, late pregnancy and immediate post-partum are quite different with high representation of secreted/extracellular matrix-related genes. Moreover, pathway-oriented ontological analysis shows that metabolism regulated by cytochrome P450 and chemokine pathways are the most significant signaling pathways regulated in late pregnancy and immediate-postpartum, respectively. Finally, increases in expression of components of the proteasome observed in both mid-pregnancy and immediate-postpartum also result in enhanced proteasome activity. Interestingly, the gene expression profiles did not correlate with the degree of cardiac hypertrophy observed in the animal groups, suggesting that distinct pathways are employed to achieve similar amounts of cardiac hypertrophy. Conclusions/Significance Our results demonstrate that cardiac adaptation to the later stages of pregnancy is quite distinct from both mid-pregnancy and exercise. Furthermore, it is very dynamic since, by 12 hours post-partum, the heart has already initiated regression of cardiac growth, and 50 genes have changed expression significantly in the immediate-postpartum compared to late-pregnancy. Thus, pregnancy-induced cardiac hypertrophy is a more complex process than exercise-induced cardiac hypertrophy and our data suggest that the mechanisms underlying the two types of hypertrophy have limited overlap.
Collapse
|
44
|
Lindsey SH, Chappell MC. Evidence that the G protein-coupled membrane receptor GPR30 contributes to the cardiovascular actions of estrogen. ACTA ACUST UNITED AC 2012; 8:343-54. [PMID: 22153880 DOI: 10.1016/j.genm.2011.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/21/2011] [Accepted: 10/26/2011] [Indexed: 02/06/2023]
Abstract
Although female protection from cardiovascular diseases declines with the fall in circulating sex hormones experienced during menopause, clinical trials in older women fail to demonstrate beneficial effects for hormone replacement therapy. The recent discovery of GPR30, a membrane-bound estrogen receptor that is structurally and functionally unique from the steroid receptors ERα and ERβ, has unveiled additional signaling pathways by which estrogen may influence cardiovascular health. This review takes an organ-based approach to assess the expression and function of GPR30 in the cardiovascular system. We concluded that although the current literature does suggest a cardiovascular role for GPR30, additional exploration is necessary to fully elucidate the estrogenic actions mediated by this novel receptor.
Collapse
Affiliation(s)
- Sarah H Lindsey
- Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157-1032, USA.
| | | |
Collapse
|
45
|
Koganti S, Snyder R, Thekkumkara T. Pharmacologic effects of 2-methoxyestradiol on angiotensin type 1 receptor down-regulation in rat liver epithelial and aortic smooth muscle cells. ACTA ACUST UNITED AC 2012; 9:76-93. [PMID: 22366193 DOI: 10.1016/j.genm.2012.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 01/04/2012] [Accepted: 01/20/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Delayed onset of cardiovascular disease (CVD) in female patients is not well understood, but could be due in part to the protective effect of estrogen before menopause. Experimental studies have identified the angiotensin type 1 receptor (AT1R) as a key factor in the progression of CVD. OBJECTIVE We examined the effects of the estrogen metabolite 2-methoxyestradiol (2ME2) on AT1R expression. METHODS Rat liver cells were exposed to 2ME2 for 24 hours, and angiotensin II (AngII) binding and AT1R mRNA expressions were assessed. RESULTS In the presence of 2ME2, cells exhibited significant down-regulation of AngII binding that was both dose and time dependent, independent of estrogen receptors (ERα/ERβ). Down-regulation of AngII binding was AT1R specific, with no change in receptor affinity. Under similar conditions, we observed lower expression of AT1R mRNA, significant inhibition of AngII-mediated increase in intracellular Ca(2+), and increased phosphorylation of ERK1/2. Pretreatment of cells with the MEK inhibitor PD98059 prevented 2ME2-induced ERK1/2 phosphorylation and down-regulation of AT1R expression, which suggests that the observed inhibitory effect is mediated through ERK1/2 signaling intermediates. Similar analyses in stably transfected CHO (Chinese hamster ovary) cell lines with a constitutively active cytomegalovirus promoter showed no change in AT1R expression, which suggests that 2ME2-mediated effects are through transcriptional regulation. The effects of 2ME2 on AT1R down-regulation through ERK1/2 were consistently reproduced in primary rat aortic smooth muscle cells. CONCLUSIONS Because AT1R has a critical role in the control of CVD, 2ME2-induced changes in receptor expression may provide beneficial effects to the cardiovascular and other systems.
Collapse
MESH Headings
- 2-Methoxyestradiol
- Angiotensin II/drug effects
- Angiotensin II/genetics
- Angiotensin II/metabolism
- Animals
- Aorta/drug effects
- Cricetinae
- Down-Regulation
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Female
- Gene Expression Regulation/drug effects
- Humans
- Liver/drug effects
- Male
- Microscopy, Fluorescence
- Myocytes, Smooth Muscle/drug effects
- RNA, Messenger/metabolism
- Rats
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Sivaramakrishna Koganti
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | | |
Collapse
|
46
|
Wang H, Jessup JA, Lin MS, Chagas C, Lindsey SH, Groban L. Activation of GPR30 attenuates diastolic dysfunction and left ventricle remodelling in oophorectomized mRen2.Lewis rats. Cardiovasc Res 2012; 94:96-104. [PMID: 22328091 DOI: 10.1093/cvr/cvs090] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS GPR30 is a novel oestrogen receptor expressed in various tissues, including the heart. We determined the role of GPR30 in the maintenance of left ventricular (LV) structure and diastolic function after the surgical loss of ovarian hormones in the female mRen2.Lewis rat, a model emulating the cardiac phenotype of the post-menopausal woman. METHODS AND RESULTS Bilateral oophorectomy (OVX) or sham surgery was performed in study rats; the selective GPR30 agonist, G-1 (50 µg/kg/day), or vehicle was given subcutaneously to OVX rats from 13-15 weeks of age. Similar to the cardiac phenotype of sham rats, G-1 preserved diastolic function and structure relative to vehicle-treated OVX littermates independent of changes in blood pressure. G-1 limited the OVX-induced increase in LV filling pressure, LV mass, wall thickness, interstitial collagen deposition, atrial natriuretic factor and brain natriuretic peptide mRNA levels, and cardiac NAD(P)H oxidase 4 (NOX4) expression. In vitro studies showed that G-1 inhibited angiotensin II-induced hypertrophy in H9c2 cardiomyocytes, evidenced by reductions in cell size, protein content per cell, and atrial natriuretic factor mRNA levels. The GPR30 antagonist, G15, inhibited the protective effects of both oestradiol and G-1 on this hypertrophy. CONCLUSION These data show that the GPR30 agonist G-1 mitigates the adverse effects of oestrogen loss on LV remodelling and the development of diastolic dysfunction in the study rats. This expands our knowledge of the sex-specific mechanisms underlying diastolic dysfunction and provides a potential therapeutic target for reducing the progression of this cardiovascular disease process in post-menopausal women.
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, Medical Center Boulevard, Winston-Salem, NC 27157-1009, USA
| | | | | | | | | | | |
Collapse
|
47
|
Hewitson TD, Zhao C, Wigg B, Lee SW, Simpson ER, Boon WC, Samuel CS. Relaxin and castration in male mice protect from, but testosterone exacerbates, age-related cardiac and renal fibrosis, whereas estrogens are an independent determinant of organ size. Endocrinology 2012; 153:188-99. [PMID: 22028442 DOI: 10.1210/en.2011-1311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study determined the effects of castration and hormone replacement therapy on the age-related cardiac and renal pathology of male relaxin gene-knockout (RlnKO) and age-matched wild-type (RlnWT) mice and that of aged male aromatase knockout (ArKO) mice, which lack estrogens and have 5-10 times the androgen levels of male wild-type mice. One-month-old RlnWT and RlnKO mice were bilaterally gonadectomized or sham operated and maintained until 12 months. Subgroups of castrated animals received testosterone or 17β-estradiol treatment from 9 to 12 months. Male ArKO mice and aromatase wild-type mice were aged to 12 months. Collected heart and kidney tissues were assessed for changes in organ size and fibrosis. Castration reduced body, heart, left ventricle, and kidney weights in both RlnKO and RlnWT mice, and the cardiac/renal fibrosis that was seen in sham RlnKO animals (all P < 0.05 vs. respective sham). Testosterone normalized organ weights and organ weight to body weight ratio of castrated animals and increased cardiac/renal collagen concentration to levels measured in or beyond that of sham RlnKO mice (all P < 0.05 vs. respective castrated mice). Furthermore, expression of TGF-β1, mothers against decapentaplegic homolog 2 (Smad2), and myofibroblast differentiation paralleled the above changes (all P < 0.05 vs. respective castrated mice), whereas matrix metalloproteinase-13 was decreased in testosterone-treated RlnKO mice. Conversely, 17β-estradiol only restored changes in organ size. Consistent with these findings, intact ArKO mice demonstrated increased cardiac/renal fibrosis in the absence of changes in organ size. These findings suggest that relaxin and castration protect, whereas androgens exacerbate, cardiac and renal fibrosis during ageing, whereas estrogens, in synergy with relaxin, regulates age-related changes in organ size.
Collapse
Affiliation(s)
- Tim D Hewitson
- Howard Florey Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Puzianowska-Kuźnicka M. ESR1 in myocardial infarction. Clin Chim Acta 2012; 413:81-7. [DOI: 10.1016/j.cca.2011.10.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/19/2011] [Accepted: 10/20/2011] [Indexed: 11/17/2022]
|
49
|
Mitsui T, Ishida M, Izawa M, Kagami Y, Arita J. Inhibition of Bcl3 gene expression mediates the anti-proliferative action of estrogen in pituitary lactotrophs in primary culture. Mol Cell Endocrinol 2011; 345:68-78. [PMID: 21787835 DOI: 10.1016/j.mce.2011.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 06/09/2011] [Accepted: 07/08/2011] [Indexed: 01/13/2023]
Abstract
In addition to their well-known stimulatory action, estrogens have an anti-proliferative effect. The present study was undertaken to investigate the mechanism by which 17β-estradiol (E2) inhibits insulin-like growth factor-1 (IGF-1)-induced proliferation in vitro in the rat pituitary lactotroph, a typical estrogen-responsive cell. E2 treatment of pituitary cells did not change levels of IGF-1-induced phosphorylation of proliferation-related protein kinases such as Erk1/2 and Akt. We performed global gene expression profiling by DNA microarray analysis and identified 177 genes regulated by E2 in the presence of IGF-1. These results were verified by quantitative real time PCR. The estrogen-regulated genes included several NFκB family related genes. As pharmacological inhibition of the NFκB pathway blocked IGF-1-induced lactotroph proliferation, we chose to investigate whether one NFκB pathway gene, Bcl3, was involved in the anti-proliferative action of E2. RNA interference-mediated knockdown of Bcl3 expression attenuated IGF-1-induced lactotroph proliferation. Even minimal induced overexpression of Bcl3 blocked the anti-proliferative action of E2. In contrast, Nfkb2, another E2-downregulated protein, required maximal overexpression to block the anti-proliferative action of E2. These results suggest that inhibition of Bcl3 expression is involved in the anti-proliferative action of estrogens in pituitary lactotrophs in culture.
Collapse
Affiliation(s)
- Tetsuo Mitsui
- Department of Physiology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | | | | | | | | |
Collapse
|
50
|
Caruso S, Cianci A, Malandrino C, Cavallari L, Gambadoro O, Arena G, Pispisa L, Agnello C, Romano M, Cavallari V. Ultrastructural and Quantitative Study of Clitoral Cavernous Tissue from Living Subjects. J Sex Med 2011; 8:1675-85. [DOI: 10.1111/j.1743-6109.2011.02253.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|