1
|
Doul J, Minaříková M, Charvátová Z, Maxová H. Nitric oxide is involved in the cardioprotection of neonatal rat hearts, but not in neonatal ischemic postconditioning. Physiol Rep 2024; 12:e16147. [PMID: 39097984 PMCID: PMC11298247 DOI: 10.14814/phy2.16147] [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: 02/17/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The cardioprotective effect of ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) in adult hearts is mediated by nitric oxide (NO). During the early developmental period, rat hearts exhibit higher resistance to ischemia-reperfusion (I/R) injury, contain higher levels of serum nitrates, and their resistance cannot be further increased by IPC or IPoC. NOS blocker (L-NAME) lowers their high resistance. Wistar rat hearts (postnatal Days 1 and 10) were perfused according to Langendorff and exposed to 40 min of global ischemia followed by reperfusion with or without IPoC. NO and reactive oxygen species donors (DEA-NONO, SIN-1) and L-NAME were administered. Tolerance to ischemia decreased between Days 1 and 10. DEA-NONO (low concentrations) significantly increased tolerance to I/R injury on both Days 1 and 10. SIN-1 increased tolerance to I/R injury on Day 10, but not on Day 1. L-NAME significantly reduced resistance to I/R injury on Day 1, but actually increased resistance to I/R injury on Day 10. Cardioprotection by IPoC on Day 10 was not affected by either NO donors or L-NAME. It can be concluded that resistance of the neonatal heart to I/R injury is NO dependent, but unlike in adult hearts, cardioprotective interventions, such as IPoC, are most likely NO independent.
Collapse
Affiliation(s)
- Jan Doul
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Marcela Minaříková
- Department of Physiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Zuzana Charvátová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Hana Maxová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
- Center for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
2
|
Wang Z, Zhang G, Hu S, Fu M, Zhang P, Zhang K, Hao L, Chen S. Research progress on the protective effect of hormones and hormone drugs in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 176:116764. [PMID: 38805965 DOI: 10.1016/j.biopha.2024.116764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Ischemic heart disease (IHD) is a condition where the heart muscle does not receive enough blood flow, leading to cardiac dysfunction. Restoring blood flow to the coronary artery is an effective clinical therapy for myocardial ischemia. This strategy helps lower the size of the myocardial infarction and improves the prognosis of patients. Nevertheless, if the disrupted blood flow to the heart muscle is restored within a specific timeframe, it leads to more severe harm to the previously deprived heart tissue. This condition is referred to as myocardial ischemia/reperfusion injury (MIRI). Until now, there is a dearth of efficacious strategies to prevent and manage MIRI. Hormones are specialized substances that are produced directly into the circulation by endocrine organs or tissues in humans and animals, and they have particular effects on the body. Hormonal medications utilize human or animal hormones as their active components, encompassing sex hormones, adrenaline medications, thyroid hormone medications, and others. While several studies have examined the preventive properties of different endocrine hormones, such as estrogen and hormone analogs, on myocardial injury caused by ischemia-reperfusion, there are other hormone analogs whose mechanisms of action remain unexplained and whose safety cannot be assured. The current study is on hormones and hormone medications, elucidating the mechanism of hormone pharmaceuticals and emphasizing the cardioprotective effects of different endocrine hormones. It aims to provide guidance for the therapeutic use of drugs and offer direction for the examination of MIRI in clinical therapy.
Collapse
Affiliation(s)
- Zhongyi Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Gaojiang Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Meilin Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Pingyuan Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kuo Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Sichong Chen
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
3
|
Hubacher V, Egle M, Graf S, Arnold M, Segiser A, Sanz MN, Casoni D, Garcia Casalta L, Nettelbeck K, Mihalj M, Siepe M, Kadner A, Longnus S. Open- vs. closed-chest pig models of donation after circulatory death. Front Cardiovasc Med 2024; 11:1325160. [PMID: 38938649 PMCID: PMC11210375 DOI: 10.3389/fcvm.2024.1325160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
Background During donation after circulatory death (DCD), cardiac grafts are exposed to potentially damaging conditions that can impact their quality and post-transplantation outcomes. In a clinical DCD setting, patients have closed chests in most cases, while many experimental models have used open-chest conditions. We therefore aimed to investigate and characterize differences in open- vs. closed-chest porcine models. Methods Withdrawal of life-sustaining therapy (WLST) was simulated in anesthetized juvenile male pigs by stopping mechanical ventilation following the administration of a neuromuscular block. Functional warm ischemic time (fWIT) was defined to start when systolic arterial pressure was <50 mmHg. Hemodynamic changes and blood chemistry were analyzed. Two experimental groups were compared: (i) an open-chest group with sternotomy prior to WLST and (ii) a closed-chest group with sternotomy after fWIT. Results Hemodynamic changes during the progression from WLST to fWIT were initiated by a rapid decline in blood oxygen saturation and a subsequent cardiovascular hyperdynamic (HD) period characterized by temporary elevations in heart rates and arterial pressures in both groups. Subsequently, heart rate and systolic arterial pressure decreased until fWIT was reached. Pigs in the open-chest group displayed a more rapid transition to the HD phase after WLST, with peak heart rate and peak rate-pressure product occurring significantly earlier. Furthermore, the HD phase duration tended to be shorter and less intense (lower peak rate-pressure product) in the open-chest group than in the closed-chest group. Discussion Progression from WLST to fWIT was more rapid, and the hemodynamic changes tended to be less pronounced in the open-chest group than in the closed-chest group. Our findings support clear differences between open- and closed-chest models of DCD. Therefore, recommendations for clinical DCD protocols based on findings in open-chest models must be interpreted with care.
Collapse
Affiliation(s)
- Valentin Hubacher
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Manuel Egle
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Selianne Graf
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Maria Arnold
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Adrian Segiser
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Maria Nieves Sanz
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Daniela Casoni
- Experimental Surgery Facility (ESF), Experimental Animal Center, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Luisana Garcia Casalta
- Experimental Surgery Facility (ESF), Experimental Animal Center, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Kay Nettelbeck
- Experimental Surgery Facility (ESF), Experimental Animal Center, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Maks Mihalj
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Advanced Cardiopulmonary Therapies and Transplantation, University of Texas Health Science Center at Houston, Texas Medical Center, Houston, TX, United States
| | - Matthias Siepe
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alexander Kadner
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sarah Longnus
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Ostadal B, Kolar F. Sixty Years of Heart Research in the Institute of Physiology of the Czech Academy of Sciences. Physiol Res 2024; 73:S35-S48. [PMID: 38634652 PMCID: PMC11412335 DOI: 10.33549/physiolres.935337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
In 2023, six decades have elapsed since the first experimental work on the heart muscle was published, in which a member of the Institute of Physiology of the Czech Academy of Sciences participated as an author; Professor Otakar Poupa was the founder and protagonist of this research domain. Sixty years - more than half of the century - is certainly significant enough anniversary that is worth looking back and reflecting on what was achieved during sometimes very complicated periods of life. It represents the history of an entire generation of experimental cardiologists; it is possible to learn from its successes and mistakes. The objective of this review is to succinctly illuminate the scientific trajectory of an experimental cardiological department over a 60-year span, from its inaugural publication to the present. The old truth - historia magistra vitae - is still valid. Keywords: Heart, Adaptation, Development, Hypoxia, Protection.
Collapse
Affiliation(s)
- B Ostadal
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | |
Collapse
|
5
|
Martin F, Xiao Y, Welten V, Nakamori K, Gizlenci M, Zhou H, Tullius SG. The combinatorial effect of age and biological sex on alloimmunity and transplantation outcome. FRONTIERS IN TRANSPLANTATION 2024; 2:1325232. [PMID: 38993871 PMCID: PMC11235293 DOI: 10.3389/frtra.2023.1325232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 07/13/2024]
Abstract
Both age and biological sex affect transplantation outcomes. We have recently shown in a large volume clinical analysis utilizing the SRTR data that graft survival is inferior in young female kidney transplant recipients. In this multi-factorial analysis, older female recipients presented with a trend towards improved transplant outcomes compared to both young female recipients and male recipients of any age. Those data supported by reports of those of others suggest that sex and age impact alloimmune responses both, individually and synergistically. Biological sex and hormone levels change throughout a lifetime with recognized effects on longevity in addition to an impact on the development and course of several disease preconditions. Detailed mechanisms of those sex and age-specific aspects have thus far been studied outside of transplantation. Effects on alloimmunity are largely unknown. Moreover, the combinatorial impact that both, biological sex and age have on transplant outcomes is not understood. Here, we summarize available data that analyze how age in combination with biological sex may shape alloimmune responses and affect transplant outcomes.
Collapse
Affiliation(s)
- Friederike Martin
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Surgery, Campus Charité Mitte|Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Yao Xiao
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Vanessa Welten
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Keita Nakamori
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Merih Gizlenci
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hao Zhou
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Liu Z, Sammani S, Barber CJ, Kempf CL, Li F, Yang Z, Bermudez RT, Camp SM, Herndon VR, Furenlid LR, Martin DR, Garcia JGN. An eNAMPT-neutralizing mAb reduces post-infarct myocardial fibrosis and left ventricular dysfunction. Biomed Pharmacother 2024; 170:116103. [PMID: 38160623 PMCID: PMC10872269 DOI: 10.1016/j.biopha.2023.116103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
Myocardial infarction (MI) triggers adverse ventricular remodeling (VR), cardiac fibrosis, and subsequent heart failure. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) is postulated to play a significant role in VR processing via activation of the TLR4 inflammatory pathway. We hypothesized that an eNAMPT specific monoclonal antibody (mAb) could target and neutralize overexpressed eNAMPT post-MI and attenuate chronic cardiac inflammation and fibrosis. We investigated humanized ALT-100 and ALT-300 mAb with high eNAMPT-neutralizing capacity in an infarct rat model to test our hypothesis. ALT-300 was 99mTc-labeled to generate 99mTc-ALT-300 for imaging myocardial eNAMPT expression at 2 hours, 1 week, and 4 weeks post-IRI. The eNAMPT-neutralizing ALT-100 mAb (0.4 mg/kg) or saline was administered intraperitoneally at 1 hour and 24 hours post-reperfusion and twice a week for 4 weeks. Cardiac function changes were determined by echocardiography at 3 days and 4 weeks post-IRI. 99mTc-ALT-300 uptake was initially localized to the ischemic area at risk (IAR) of the left ventricle (LV) and subsequently extended to adjacent non-ischemic areas 2 hours to 4 weeks post-IRI. Radioactive uptake (%ID/g) of 99mTc-ALT-300 in the IAR increased from 1 week to 4 weeks (0.54 ± 0.16 vs. 0.78 ± 0.13, P < 0.01). Rats receiving ALT-100 mAb exhibited significantly improved myocardial histopathology and cardiac function at 4 weeks, with a significant reduction in the collagen volume fraction (%LV) compared to controls (21.5 ± 6.1% vs. 29.5 ± 9.9%, P < 0.05). Neutralization of the eNAMPT/TLR4 inflammatory cascade is a promising therapeutic strategy for MI by reducing chronic inflammation, fibrosis, and preserving cardiac function.
Collapse
Affiliation(s)
- Zhonglin Liu
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States; Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States.
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Christy J Barber
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L Kempf
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Feng Li
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Zhen Yang
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Rosendo T Bermudez
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M Camp
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Vivian Reyes Herndon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Diego R Martin
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States.
| | - Joe G N Garcia
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| |
Collapse
|
7
|
Al-Katat A, Bergeron A, Parent L, Lorenzini M, Fiset C, Calderone A. Rapamycin treatment unmasks a sex-specific pattern of scar expansion of the infarcted rat heart: The relationship between mTOR and K ATP channel. IUBMB Life 2023; 75:717-731. [PMID: 36988388 DOI: 10.1002/iub.2722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/24/2023] [Indexed: 03/30/2023]
Abstract
Inhibition of the mammalian target of rapamycin (mTOR) with the macrolide rapamycin or pharmacological suppression of KATP channel opening translated to scar expansion of the myocardial infarcted (MI) adult female rodent heart. The present study tested the hypotheses that rapamycin-mediated scar expansion was sex-specific and that mTOR signaling directly influenced KATP channel subunit expression/activity. Scar size was significantly larger in post-MI male rats as compared to the previous data reported in post-MI female rats. The reported scar expansion of rapamycin-treated post-MI female rats was not observed following the administration of the macrolide to post-MI male rats. Protein levels of the KATP channel subunits Kir6.2 and SUR2A and phosphorylation of the serine2448 residue of mTOR were similar in the normal heart of adult male and female rats. By contrast, greater tuberin inactivation characterized by the increased phosphorylation of the threonine1462 residue and reduced raptor protein levels were identified in the normal heart of adult female rats. Rapamycin pretreatment of phorbol 12,13-dibutyrate (PDBu)-treated neonatal rat ventricular cardiomyocytes (NNVMs) suppressed hypertrophy, inhibited p70S6K phosphorylation, and attenuated SUR2A protein upregulation. In the presence of low ATP levels, KATP channel activity detected in untreated NNVMs was significantly attenuated in PDBu-induced hypertrophied NNVMs via a rapamycin-independent pathway. Thus, rapamycin administration to post-MI rats unmasked a sex-specific pattern of scar expansion and mTOR signaling in PDBu-induced hypertrophied NNVMs significantly increased SUR2A protein levels. However, the biological advantage associated with SUR2A protein upregulation was partially offset by an mTOR-independent pathway that attenuated KATP channel activity in PDBu-induced hypertrophied NNVMs.
Collapse
Affiliation(s)
- Aya Al-Katat
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada
| | - Alexandre Bergeron
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Lucie Parent
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Lorenzini
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Celine Fiset
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Montréal, Québec, Canada
| | - Angelino Calderone
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
8
|
Sabe SA, Harris DD, Broadwin M, Sabra M, Xu CM, Banerjee D, Abid MR, Sellke FW. Sitagliptin therapy improves myocardial perfusion and arteriolar collateralization in chronically ischemic myocardium: A pilot study. Physiol Rep 2023; 11:e15744. [PMID: 37300400 PMCID: PMC10257079 DOI: 10.14814/phy2.15744] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
Dipeptidyl peptidase 4 inhibitors (DPP4i) may be cardioprotective based on several small animal and clinical studies, though randomized control trials have demonstrated limited benefit. Given these discrepant findings, the role of these agents in chronic myocardial disease, particularly in the absence of diabetes, is still poorly understood. The purpose of this study was to determine the effects of sitagliptin, a DPP4i, on myocardial perfusion and microvessel density in a clinically relevant large animal model of chronic myocardial ischemia. Normoglycemic Yorkshire swine underwent ameroid constrictor placement to the left circumflex artery to induce chronic myocardial ischemia. Two weeks later, pigs received either no drug (CON, n = 8) or 100 mg oral sitagliptin (SIT) daily (n = 5). Treatment continued for 5 weeks, followed by hemodynamic measurements, euthanasia, and tissue harvest of ischemic myocardium. There were no significant differences in myocardial function between CON and SIT as measured by stroke work (p > 0.5), cardiac output (p = 0.22), and end-systolic elastance (p = 0.17). SIT was associated with increased absolute blood flow at rest (17% increase, IQR 12-62, p = 0.045) and during pacing (89% increase, IQR 83-105, p = 0.002). SIT was also associated with improved arteriolar density (p = 0.045) compared with CON, without changes in capillary density (p = 0.72). SIT was associated with increased expression of pro-arteriogenic markers MCP-1 (p = 0.003), TGFß (p = 0.03), FGFR1 (p = 0.002), and ICAM-1 (p = 0.03), with a trend toward an increase in the ratio of phosphorylated/active PLCγ1 to total PLCγ1 (p = 0.11) compared with CON. In conclusion, in chronically ischemic myocardium, sitagliptin improves myocardial perfusion and arteriolar collateralization via the activation of pro-arteriogenic signaling pathways.
Collapse
Affiliation(s)
- Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Dwight Douglas Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Cynthia M. Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Debolina Banerjee
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island HospitalAlpert Medical School of Brown University, Rhode Island HospitalProvidenceRhode IslandUSA
| |
Collapse
|
9
|
Heger J, Szabados T, Brosinsky P, Bencsik P, Ferdinandy P, Schulz R. Sex Difference in Cardioprotection against Acute Myocardial Infarction in MAO-B Knockout Mice In Vivo. Int J Mol Sci 2023; 24:ijms24076443. [PMID: 37047416 PMCID: PMC10094730 DOI: 10.3390/ijms24076443] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
The cardiomyocyte-specific knockout (KO) of monoamine oxidase (MAO)-B, an enzyme involved in the formation of reactive oxygen species (ROS), reduced myocardial ischemia/reperfusion (I/R) injury in vitro. Because sex hormones have a strong impact on MAO metabolic pathways, we analyzed the myocardial infarct size (IS) following I/R in female and male MAO-B KO mice in vivo. Method and Results: To induce the deletion of MAO-B, MAO-B KO mice (Myh6 Cre+/MAO-Bfl/fl) and wild-type (WT, Cre-negative MAO-Bfl/fl littermates) were fed with tamoxifen for 2 weeks followed by 10 weeks of normal mice chow. Myocardial infarction (assessed by TTC staining and expressed as a percentage of the area at risk as determined by Evans blue staining)) was induced by 45 min coronary occlusion followed by 120 min of reperfusion. Results: The mortality following I/R was higher in male compared to female mice, with the lowest mortality found in MAO-B KO female mice. IS was significantly higher in male WT mice compared to female WT mice. MAO-B KO reduced IS in male mice but had no further impact on IS in female MAO-B KO mice. Interestingly, there was no difference in the plasma estradiol levels among the groups. Conclusion: The cardiomyocyte-specific knockout of MAO-B protects male mice against acute myocardial infarction but had no effect on the infarct size in female mice.
Collapse
|
10
|
Ishiwata-Endo H, Kato J, Oda H, Sun J, Yu ZX, Liu C, Springer DA, Dagur P, Lizak MJ, Murphy E, Moss J. Mono-ADP-ribosyltransferase 1 ( Artc1 )-deficiency decreases tumorigenesis, increases inflammation, decreases cardiac contractility, and reduces survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527366. [PMID: 36945646 PMCID: PMC10028742 DOI: 10.1101/2023.02.06.527366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arginine-specific mono-ADP-ribosylation is a reversible post-translational modification; arginine-specific, cholera toxin-like mono-ADP-ribosyltransferases (ARTCs) transfer ADP-ribose from NAD + to arginine, followed by cleavage of ADP-ribose-(arginine)protein bond by ADP-ribosylarginine hydrolase 1 (ARH1), generating unmodified (arginine)protein. ARTC1 has been shown to enhance tumorigenicity as does Arh1 deficiency. In this study, Artc1 -KO and Artc1/Arh1 -double-KO mice showed decreased spontaneous tumorigenesis and increased age-dependent, multi-organ inflammation with upregulation of pro-inflammatory cytokine TNF- α . In a xenograft model using tumorigenic Arh1 -KO mouse embryonic fibroblasts (MEFs), tumorigenicity was decreased in Artc1 -KO and heterozygous recipient mice, with tumor infiltration by CD8 + T cells and macrophages, leading to necroptosis, suggesting that ARTC1 promotes the tumor microenvironment. Furthermore, Artc1/Arh1 -double-KO MEFs showed decreased tumorigenesis in nude mice, showing that tumor cells as well as tumor microenvironment require ARTC1. By echocardiography and MRI, Artc1 -KO and heterozygous mice showed male-specific, reduced myocardial contractility. Furthermore, Artc1 -KO male hearts exhibited enhanced susceptibility to myocardial ischemia-reperfusion-induced injury with increased receptor-interacting protein kinase 3 (RIP3) protein levels compared to WT mice, suggesting that ARTC1 suppresses necroptosis. Overall survival rate of Artc1 -KO was less than their Artc1 -WT counterparts, primarily due to enhanced immune response and inflammation. Thus, anti-ARTC1 agents may reduce tumorigenesis but may increase multi-organ inflammation and decrease cardiac contractility.
Collapse
|
11
|
Lu X, Yang B, Qi R, Xie Q, Li T, Yang J, Tong T, Niu K, Li M, Pan W, Zhang Y, Shi D, Li S, Dai C, Shen C, Wang X, Wang Y, Song J. Targeting WWP1 ameliorates cardiac ischemic injury by suppressing KLF15-ubiquitination mediated myocardial inflammation. Theranostics 2023; 13:417-437. [PMID: 36593958 PMCID: PMC9800727 DOI: 10.7150/thno.77694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Rationale: Previous studies have suggested that myocardial inflammation plays a critical role after ischemic myocardial infarction (MI); however, the underlying mechanisms still need to be fully elucidated. WW domain-containing ubiquitin E3 ligase 1 (WWP1) is considered as an important therapeutic target for cardiovascular diseases due to its crucial function in non-ischemic cardiomyopathy, though it remains unknown whether targeting WWP1 can alleviate myocardial inflammation and ischemic injury post-MI. Methods: Recombinant adeno-associated virus 9 (rAAV9)-cTnT-mediated WWP1 or Kruppel-like factor 15 (KLF15) gene transfer and a natural WWP1 inhibitor Indole-3-carbinol (I3C) were used to determine the WWP1 function in cardiomyocytes. Cardiac function, tissue injury, myocardial inflammation, and signaling changes in the left ventricular tissues were analyzed after MI. The mechanisms underlying WWP1 regulation of cardiomyocyte phenotypes in vitro were determined using the adenovirus system. Results: We found that WWP1 expression was up-regulated in cardiomyocytes located in the infarct border at the early phase of MI and in hypoxia-treated neonatal rat cardiac myocytes (NRCMs). Cardiomyocyte-specific WWP1 overexpression augmented cardiomyocyte apoptosis, increased infarct size and deteriorated cardiac function. In contrast, inhibition of WWP1 in cardiomyocytes mitigated MI-induced cardiac ischemic injury. Mechanistically, WWP1 triggered excessive cardiomyocyte inflammation after MI by targeting KLF15 to catalyze K48-linked polyubiquitination and degradation. Ultimately, WWP1-mediated degradation of KLF15 contributed to the up-regulation of p65 acetylation, and activated the inflammatory signaling of MAPK in ischemic myocardium and hypoxia-treated cardiomyocytes. Thus, targeting of WWP1 by I3C protected against cardiac dysfunction and remodeling after MI. Conclusions: Our study provides new insights into the previously unrecognized role of WWP1 in cardiomyocyte inflammation and progression of ischemic injury induced by MI. Our findings afford new therapeutic options for patients with ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Xia Lu
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Boshen Yang
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ruiqiang Qi
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China
| | - Qifei Xie
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China
| | - Taixi Li
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jie Yang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tingting Tong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Kaifan Niu
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Weijun Pan
- Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Yongxin Zhang
- The first clinical medical college, Southern Medical University, Guangzhou 510000, China
| | - Dongmei Shi
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Suiji Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaoqing Wang
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.,✉ Corresponding authors: Juan Song, E-mail: ; Yan Wang, E-mail: ; Xiaoqing Wang, E-mail:
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China.,✉ Corresponding authors: Juan Song, E-mail: ; Yan Wang, E-mail: ; Xiaoqing Wang, E-mail:
| | - Juan Song
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361004, China.,✉ Corresponding authors: Juan Song, E-mail: ; Yan Wang, E-mail: ; Xiaoqing Wang, E-mail:
| |
Collapse
|
12
|
Chen Q, Akande O, Lesnefsky EJ, Quader M. Influence of sex on global myocardial ischemia tolerance and mitochondrial function in circulatory death donor hearts. Am J Physiol Heart Circ Physiol 2023; 324:H57-H66. [PMID: 36426883 PMCID: PMC9762969 DOI: 10.1152/ajpheart.00478.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Donation after circulatory death (DCD) donor hearts are not routinely used for heart transplantation (HTx) because of ischemic damage, which is inherent to the DCD process. HTx outcomes are suboptimal in males who received female donor hearts. The exact mechanism for suboptimal outcomes from female donor hearts has not been defined. Differential susceptibility to ischemia tolerance, which would play a significant role in DCD donation, could be a reason but has not been studied. We studied the influence of sex on global myocardial ischemia tolerance and mitochondrial function. Sprague-Dawley rats of both sexes were assigned to DCD (n = 32) or control beating-heart donor (CBD, n = 28) groups. DCD hearts underwent 25 min of in vivo global myocardial ischemia and 90 min of ex vivo Krebs-Henseleit buffer perfusion at 37°C. CBD hearts were procured without ischemia. Infarct size was determined in hearts following 90 min of reperfusion, and in another set of hearts, mitochondrial function (oxidative-phosphorylation) was studied following 60 min of reperfusion. Infarct size was increased 3.3-fold in male and 3.1-fold in female DCD hearts compared with CBD hearts. However, infarct size (%) was comparable in female and male DCD hearts (male: 25.4 ± 3.7 vs. female 19.0 ± 3.3, P = NS). Oxidative phosphorylation was similarly decreased in male and female DCD hearts' mitochondria compared with CBD hearts' mitochondria. Thus, neither infarct size nor mitochondrial dysfunction was higher in female DCD hearts. These results suggest that the susceptibility to ischemia is not the reason for suboptimal HTx outcomes with female donor hearts.NEW & NOTEWORTHY The current study shows cardiac injury is not increased in female DCD hearts following global ischemia-reperfusion compared with male DCD hearts. In addition, mitochondrial dysfunction with DCD ischemia-reperfusion is comparable in both sexes. Sex-specific immune responses and hormone receptor modulation may contribute to suboptimal outcomes in male HTx recipients with female donor hearts.
Collapse
Affiliation(s)
- Qun Chen
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
- Pauley Heart Center, Virginia Commonwealth University Health System, Richmond, Virginia
| | - Oluwatoyin Akande
- Division of Cardiothoracic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
- Pauley Heart Center, Virginia Commonwealth University Health System, Richmond, Virginia
- Cardiology Section, Medical Service, McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| | - Mohammed Quader
- Pauley Heart Center, Virginia Commonwealth University Health System, Richmond, Virginia
- Division of Cardiothoracic Surgery, Virginia Commonwealth University, Richmond, Virginia
- Cardiothoracic Surgery Section, Surgical Service, McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| |
Collapse
|
13
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
14
|
Abstract
Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.
Collapse
Affiliation(s)
- Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA
- Department of Medicine, David Geffen School of Medicine at UCLA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Carrie B. Wiese
- Department of Human Genetics, David Geffen School of Medicine at UCLA
| |
Collapse
|
15
|
Chen J, Liu Y, Pan D, Xu T, Luo Y, Wu W, Wu P, Zhu H, Li D. Estrogen inhibits endoplasmic reticulum stress and ameliorates myocardial ischemia/reperfusion injury in rats by upregulating SERCA2a. Cell Commun Signal 2022; 20:38. [PMID: 35331264 PMCID: PMC8944077 DOI: 10.1186/s12964-022-00842-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background The incidence of coronary heart disease (CHD) in premenopausal women is significantly lower than that of men of the same age, suggesting protective roles of estrogen for the cardiovascular system against CHD. This study aimed to confirm the protective effect of estrogen on myocardium during myocardial ischemia/reperfusion (MI/R) injury and explore the underlying mechanisms. Methods Neonatal rat cardiomyocytes and Sprague–Dawley rats were used in this study. Different groups were treated by bilateral ovariectomy, 17β-estradiol (E2), adenoviral infection, or siRNA transfection. The expression of sarcoplasmic reticulum Ca2+ ATPase pump (SERCA2a) and endoplasmic reticulum (ER) stress-related proteins were measured in each group to examine the effect of different E2 levels and determine the relationship between SERCA2a and ER stress. The cell apoptosis, myocardial infarction size, levels of apoptosis and serum cardiac troponin I, ejection fraction, calcium transient, and morphology changes of the myocardium and ER were examined to verify the effects of E2 on the myocardium. Results Bilateral ovariectomy resulted in reduced SERCA2a levels and more severe MI/R injury. E2 treatment increased SERCA2a expression. Both E2 treatment and exogenous SERCA2a overexpression decreased levels of ER stress-related proteins and alleviated myocardial damage. In contrast, SERCA2a knockdown exacerbated ER stress and myocardial damage. Addition of E2 after SERCA2a knockdown did not effectively inhibit ER stress or reduce myocardial injury. Conclusions Our data demonstrate that estrogen inhibits ER stress and attenuates MI/R injury by upregulating SERCA2a. These results provide a new potential target for therapeutic intervention and drug discovery in CHD. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00842-2.
Collapse
Affiliation(s)
- Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Tongda Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Waqar A, Jain A, Joseph C, Srivastava K, Ochuba O, Alkayyali T, Poudel S. Cardioprotective Role of Estrogen in Takotsubo Cardiomyopathy. Cureus 2022; 14:e22845. [PMID: 35382214 PMCID: PMC8977075 DOI: 10.7759/cureus.22845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 03/04/2022] [Indexed: 11/21/2022] Open
Abstract
Takotsubo cardiomyopathy (TC) is a rare, reversible cause of left ventricular wall motion abnormality (LVWMA) that mimics the presentation of acute myocardial infarction (AMI). TC is usually preceded by an emotional or physical stressor and appears to be more common in postmenopausal women. Various pathophysiological hypotheses of TC have been proposed, but the exact mechanism of action remains elusive. Elevated levels of catecholamines leading to cardiac dysfunction are the most prevalent hypothesis. The protective role of estrogen in the development of cardiomyopathies has been studied extensively. International Takotsubo Diagnostic Criteria (InterTAK) and Mayo clinic diagnostic criteria both have the stipulation stating prevalence of TC is higher in postmenopausal women which hints towards the protective role of estrogen in the development of TC. To review the protective role of estrogen in the mechanism of this novel pathology, we searched Pubmed and Google scholar for the relevant articles by using keywords such as: “takotsubo cardiomyopathy”, “apical ballooning”, “broken heart syndrome”, “stress cardiomyopathy”, “left ventricle wall motion abnormality”, “estrogen”, “estradiol” and “sex hormones”. Our research revealed that although the prevalence of TC is greater in postmenopausal women as compared to men, the prognosis is worse in men. It also revealed the involvement of multiple cellular pathways under the influence of estrogen that could explain the cardioprotective effect of estrogen. Most of the articles found were based on animal studies, thus, there is an emphasis on future human studies. However, we strongly suggest evaluating estrogen levels as part of the initial workup for any patient presenting with signs and symptoms of cardiac pathology.
Collapse
|
17
|
Chacon-Alberty L, Ye S, Daoud D, Frankel WC, Virk H, Mase J, Hochman-Mendez C, Li M, Sampaio LC, Taylor DA, Loor G. Analysis of sex-based differences in clinical and molecular responses to ischemia reperfusion after lung transplantation. Respir Res 2021; 22:318. [PMID: 34937545 PMCID: PMC8693497 DOI: 10.1186/s12931-021-01900-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/19/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sex and hormones influence immune responses to ischemia reperfusion (IR) and could, therefore, cause sex-related differences in lung transplantation (LTx) outcomes. We compared men's and women's clinical and molecular responses to post-LTx IR. METHODS In 203 LTx patients, we used the 2016 International Society for Heart and Lung Transplantation guidelines to score primary graft dysfunction (PGD). In a subgroup of 40 patients with blood samples collected before LTx (T0) and 6, 24, 48 (T48), and 72 h (T72) after lung reperfusion, molecular response to IR was examined through serial analysis of circulating cytokine expression. RESULTS After adjustment, women had less grade 3 PGD than men at T48, but not at T72. PGD grade decreased from T0 to T72 more often in women than men. The evolution of PGD (the difference in mean PGD between T72 and T0) was greater in men. However, the evolution of IL-2, IL-7, IL-17a, and basic fibroblast growth factor levels was more often sustained throughout the 72 h in women. In the full cohort, we noted no sex differences in secondary clinical outcomes, but women had significantly lower peak lactate levels than men across the 72 h. CONCLUSIONS Men and women differ in the evolution of PGD and cytokine secretion after LTx: Women have a more sustained proinflammatory response than men despite a greater reduction in PGD over time. This interaction between cytokine and PGD responses warrants investigation. Additionally, there may be important sex-related differences that could be used to tailor treatment during or after transplantation.
Collapse
Affiliation(s)
| | - Shengbin Ye
- Department of Biostatistics, Rice University, Houston, TX, USA
| | - Daoud Daoud
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Cardiopulmonary Transplantation and Center for Cardiac Support, Texas Heart Institute, 6770 Bertner Ave, Suite 355-K, Houston, TX, 77030, USA
| | - William C Frankel
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Hassan Virk
- Department of Regenerative Medicine, Texas Heart Institute, Houston, TX, USA
- Division of Infectious Diseases, Department of Internal Medicine, Center for Antimicrobial Resistance and Microbial Genomics (CARMiG), University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jonathan Mase
- Department of Regenerative Medicine, Texas Heart Institute, Houston, TX, USA
| | | | - Meng Li
- Department of Biostatistics, Rice University, Houston, TX, USA
| | - Luiz C Sampaio
- Department of Regenerative Medicine, Texas Heart Institute, Houston, TX, USA
- Department of Advanced Cardiopulmonary Therapies and Transplantation, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Doris A Taylor
- Department of Regenerative Medicine, Texas Heart Institute, Houston, TX, USA
- RegenMedix Consulting, Houston, TX, USA
| | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
- Department of Cardiopulmonary Transplantation and Center for Cardiac Support, Texas Heart Institute, 6770 Bertner Ave, Suite 355-K, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
(Sex differences in cardiac tolerance to ischemia-reperfusion injury - the role of mitochondria). COR ET VASA 2021. [DOI: 10.33678/cor.2021.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
19
|
Zachura M, Sadowski M, Kurzawski J, Piątek K, Gąsior M. Heterogeneity of the no-reflow group after primary percutaneous coronary intervention due to ST-segment elevation myocardial infarction - are there sex differences? CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 37:97-101. [PMID: 34167912 DOI: 10.1016/j.carrev.2021.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/26/2022]
Abstract
AIMS Assessment of the diversity in the no-reflow population after primary percutaneous coronary intervention (pPCI) due to ST-segment elevation myocardial infarction (STEMI). Are there any gender-related differences? MATERIAL AND METHODS Analysis of 1063 STEMI patients with Thrombolysis in Myocardial Infarction (TIMI) grade 0 or 1 following pPCI. The study group consisted of 685 patients with TIMI grade 0 and of 378 patients with TIMI grade 1. We analyzed clinical characteristics, in-hospital mortality and 2-year follow-up in both groups. RESULTS Among women with the TIMI grade 1 an atrial fibrillation, tachycardia and impaired ejection fraction were more common than in men. The vessel responsible for myocardial infarction was most commonly the left anterior descending (LAD) in women, whereas the right coronary artery (RCA) in men. These differences were not observed in group with TIMI grade 0. We observed a higher incidence of in-hospital death in the population with TIMI grade 0 compared with TIMI grade 1 (21.9% vs 17.2%; p 0.0189). In the TIMI grade 1 group there was significantly higher incidence of in-hospital mortality in women compared to men (13.2% vs 22.7%; p 0,0159). Among women with postprocedural TIMI grade 0 in all periods of long-term follow-up the mortality was significantly higher compared to men (9.5% vs 17%; p 0,0111; 11.8% vs 19.7%; p 0.0139 and 16.7% vs 23.9%; p 0.043 for 6-,12-months and 2-years of follow up respectively). CONCLUSIONS Patients with no-reflow phenomenon in infarct related artery after pPCI constitute a more diverse group than previously thought. Some differences are most likely gender-specific. The female sex might have an adverse effect on in-hospital mortality in case of TIMI grade 1 and on the long-term prognosis among patients with TIMI grade 0.
Collapse
Affiliation(s)
- Małgorzata Zachura
- 2nd Department of Cardiology, Świętokrzyskie Cardiology Centre, Kielce, Poland; The Faculty of Medicine and Health Sciences, The Jan Kochanowski University, Kielce, Poland.
| | - Marcin Sadowski
- The Faculty of Medicine and Health Sciences, The Jan Kochanowski University, Kielce, Poland; Department of Interventional Cardiology, Świętokrzyskie Cardiology Centre, Kielce, Poland
| | - Jacek Kurzawski
- 2nd Department of Cardiology, Świętokrzyskie Cardiology Centre, Kielce, Poland
| | - Karolina Piątek
- 2nd Department of Cardiology, Świętokrzyskie Cardiology Centre, Kielce, Poland
| | - Mariusz Gąsior
- 3rd Department of Cardiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Silesian Centre for Heart Disease, Zabrze, Poland
| |
Collapse
|
20
|
Meléndez-Fernández OH, Walton JC, DeVries AC, Nelson RJ. Clocks, Rhythms, Sex, and Hearts: How Disrupted Circadian Rhythms, Time-of-Day, and Sex Influence Cardiovascular Health. Biomolecules 2021; 11:883. [PMID: 34198706 PMCID: PMC8232105 DOI: 10.3390/biom11060883] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are the top cause of mortality in the United States, and ischemic heart disease accounts for 16% of all deaths around the world. Modifiable risk factors such as diet and exercise have often been primary targets in addressing these conditions. However, mounting evidence suggests that environmental factors that disrupt physiological rhythms might contribute to the development of these diseases, as well as contribute to increasing other risk factors that are typically associated with cardiovascular disease. Exposure to light at night, transmeridian travel, and social jetlag disrupt endogenous circadian rhythms, which, in turn, alter carefully orchestrated bodily functioning, and elevate the risk of disease and injury. Research into how disrupted circadian rhythms affect physiology and behavior has begun to reveal the intricacies of how seemingly innocuous environmental and social factors have dramatic consequences on mammalian physiology and behavior. Despite the new focus on the importance of circadian rhythms, and how disrupted circadian rhythms contribute to cardiovascular diseases, many questions in this field remain unanswered. Further, neither time-of-day nor sex as a biological variable have been consistently and thoroughly taken into account in previous studies of circadian rhythm disruption and cardiovascular disease. In this review, we will first discuss biological rhythms and the master temporal regulator that controls these rhythms, focusing on the cardiovascular system, its rhythms, and the pathology associated with its disruption, while emphasizing the importance of the time-of-day as a variable that directly affects outcomes in controlled studies, and how temporal data will inform clinical practice and influence personalized medicine. Finally, we will discuss evidence supporting the existence of sex differences in cardiovascular function and outcomes following an injury, and highlight the need for consistent inclusion of both sexes in studies that aim to understand cardiovascular function and improve cardiovascular health.
Collapse
Affiliation(s)
- O. Hecmarie Meléndez-Fernández
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| | - A. Courtney DeVries
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV 26505, USA;
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (J.C.W.); (R.J.N.)
| |
Collapse
|
21
|
Shi H, Gao Y, Dong Z, Yang J, Gao R, Li X, Zhang S, Ma L, Sun X, Wang Z, Zhang F, Hu K, Sun A, Ge J. GSDMD-Mediated Cardiomyocyte Pyroptosis Promotes Myocardial I/R Injury. Circ Res 2021; 129:383-396. [PMID: 34015941 PMCID: PMC8291144 DOI: 10.1161/circresaha.120.318629] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Pyroptosis is a morphologically and mechanistically distinct form of cell death and is characterized by GSDMD (gasdermin D) or GSDME (gasdermin E)-mediated necrosis with excessive inflammatory factor release. Cardiomyocyte necrosis and inflammation play key roles in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury. However, whether cardiomyocytes undergo pyroptosis and the underlying mechanism in myocardial I/R injury remain unclear. Objective: We aimed to investigate the role of pyroptosis in myocardial I/R injury. Methods and Results: In vivo and in vitro experiments were used to investigate pyroptosis of cardiomyocyte and the associated mechanisms during I/R injury. Wild-type, Myh6-Cre, and cardiomyocyte-specific GSDMD-deficient male mice were subjected to I/R. Human peripheral blood samples were collected from patients with acute ST-segment–elevation myocardial infarction or control patients at 0, 1, and 24 hours after percutaneous coronary intervention in our department. The serum levels of GSDMD were measured by ELISA. Hypoxia/reoxygenation induced cardiomyocyte pyroptosis and the release of mature IL (interleukin)-18 but not IL-1β, which mechanistically resulted from GSDMD cleavage by caspase-11 in cardiomyocytes. Furthermore, GSDMD gene deletion blocked hypoxia/reoxygenation-induced cardiomyocyte pyroptosis and IL-18 release. GSDMD and its pyroptosis-inducing N-terminal fragment were upregulated in myocardial tissues after I/R injury. Immunofluorescence analysis showed that GSDMD was mainly localized in cardiomyocytes. GSDMD deficiency in cardiomyocytes significantly reduced the I/R-induced myocardial infarct size. Moreover, increased GSDMD serum levels were detected in patients exhibiting I/R injury 1 hour after percutaneous coronary intervention for ST-segment–elevation myocardial infarction. Conclusions: Our results show that GSDMD-mediated cardiomyocyte pyroptosis is a key event during myocardial I/R injury and that the caspase-11/GSDMD pathway may be essential to this process. Additionally, GSDMD inhibition significantly reduces cardiomyocyte pyroptosis and I/R-induced myocardial injury. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Huairui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Zhen Dong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Ji'e Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Rifeng Gao
- Shanghai Fifth Peoples Hospital, Fudan University, Shanghai, China (R.G.)
| | - Xiao Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Shuqi Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Leilei Ma
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Xiaolei Sun
- Institute of Biomedical Science, Fudan University, Shanghai, China (X.S., Z.W., A.S., J.G.)
| | - Zeng Wang
- Institute of Biomedical Science, Fudan University, Shanghai, China (X.S., Z.W., A.S., J.G.)
| | - Feng Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Kai Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Aijun Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Institute of Biomedical Science, Fudan University, Shanghai, China (X.S., Z.W., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Institute of Biomedical Science, Fudan University, Shanghai, China (X.S., Z.W., A.S., J.G.).,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China (H.S., Y.G., Z.D., J.Y., X.L., S.Z., L.M., F.Z., K.H., A.S., J.G.)
| |
Collapse
|
22
|
Lassen TR, Hjortbak MV, Hauerslev M, Tonnesen PT, Kristiansen SB, Jensen RV, Bøtker HE. Influence of strain, age, origin, and anesthesia on the cardioprotective efficacy by local and remote ischemic conditioning in an ex vivo rat model. Physiol Rep 2021; 9:e14810. [PMID: 33818005 PMCID: PMC8020046 DOI: 10.14814/phy2.14810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background Local ischemic preconditioning (IPC) and remote ischemic conditioning (RIC) induced by brief periods of ischemia and reperfusion protect against ischemia‐reperfusion injury. Methods We studied the sensitivity to IR‐injury and the influence of strain, age, supplier, and anesthesia upon the efficacy of IPC and RIC in 7‐ and 16‐weeks‐old Sprague‐Dawley and Wistar rats from three different suppliers. The influence of sedation with a hypnorm and midazolam mixture (rodent mixture) and pentobarbiturate was compared. Results IPC attenuated infarct size in both 7‐weeks‐old Sprague–Dawley (48.4 ± 17.7% vs. 20.3 ± 6.9, p < 0.001) and 7‐weeks‐old Wistar (55.6 ± 10.9% vs. 26.8 ± 5.0%, p < 0.001) rats. Infarct size was larger in 16‐weeks‐old Sprague–Dawley rats, however, IPC still lowered infarct size (78.8 ± 9.2% vs. 58.3 ± 12.3%, p < 0.01). RIC reduced infarct sizes in 7‐weeks‐old Sprague–Dawley (75.3 ± 11.8% vs. 58.6 ± 8.9%, p < 0.05), but not in 7‐weeks‐old Wistar rats (31.7 ± 17.6% and 24.0 ± 12.6%, p = 0.2). In 16‐weeks‐old Sprague–Dawley rats, RIC did not induce protection (76.4 ± 5.5% and 73.2 ± 14.7%, p = 0.6). However, RIC induced protection in 16‐weeks‐old Wistar rats (45.2 ± 8.5% vs. 14.7 ± 10.8%, p < 0.001). RIC did not reduce infarct size in 7‐weeks‐old Sprague–Dawley rats from Charles River (62.0 ± 13.5% and 69.4 ± 10.4% p = 0.3) or 16‐weeks‐old Wistar rats from Janvier (50.7 ± 11.3 and 49.2 ± 16.2, p = 0.8). There was no difference between sedation with rodent mixture or pentobarbiturate. Conclusion The cardioprotective effect of IPC is consistent across rat strains independent of age, strain, and supplier. RIC seems to be less reproducible, but still yields protection across different rat strains. However, age, animal supplier, and anesthetics may modulate the sensitivity of IR‐injury and the response to RIC.
Collapse
Affiliation(s)
- Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Hauerslev
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Pernille Tilma Tonnesen
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
23
|
Zhou Y, Peng W, Yang G, Pan X, Ding N, Zhang H, Peng Z, Zhang D, Wu S, Chai X. Gender Difference is Associated with Short-Term Outcomes in Non-Surgically Managed Acute Aortic Dissection Patients with Hypertension: A Retrospective Cohort Study. Risk Manag Healthc Policy 2021; 14:323-330. [PMID: 33536801 PMCID: PMC7850566 DOI: 10.2147/rmhp.s289943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background The management of acute aortic dissection (AAD) has improved; however, the outcomes related to different gender with short-term outcomes in non-surgically managed AAD with hypertension are still limited. Our objective was to explore gender-differences in association with short-term outcomes of patients comorbid with hypertension in non-surgically managed AAD. Methods This is an observational retrospective single-center cohort. We analyzed the data from the Second Xiangya Hospital of Central South University (2014-2018). The data on demographics, clinical presentation, chronic comorbidities, laboratory testing, imaging studies, and treatment were analyzed for all patients. Univariate and multiple analyses were used to test gender-difference associated with short-term outcomes of patients with hypertension in non-surgically managed AAD. Results In total, 288 patients were enrolled in this study, of whom 238 (82.63%) were male and 50 (17.37%) were females. About 74% of female patients were dead in-hospital, which was more than male patients (56.3%). Female patients with diabetes mellitus were more than male patients (14% vs 2.94%), while male patients with smoking were significant higher than female patients (36.55% vs 8%). In the full model (model 3), after adjusting for confounding variables, the female AAD patients were more likely to have worse short-term outcomes (OR=3.60, 95% CI=1.41 to 9.60). Conclusion Female patients were more likely to have worse outcomes in non-surgically managed AAD patients with hypertension. Large numbers of investigations are required to further explore this relationship.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Wen Peng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Xiaogao Pan
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Ning Ding
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Hongliang Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Zhenyu Peng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Dongshan Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| | - Sijie Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People's Republic of China
| |
Collapse
|
24
|
Manning JR, Wijeratne AB, Oloizia BB, Zhang Y, Greis KD, Schultz JEJ. Phosphoproteomic analysis identifies phospho-Threonine-17 site of phospholamban important in low molecular weight isoform of fibroblast growth factor 2-induced protection against post-ischemic cardiac dysfunction. J Mol Cell Cardiol 2020; 148:1-14. [PMID: 32853649 DOI: 10.1016/j.yjmcc.2020.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/04/2020] [Accepted: 08/09/2020] [Indexed: 10/23/2022]
Abstract
RATIONALE Among its many biological roles, fibroblast growth factor 2 (FGF2) protects the heart from dysfunction and damage associated with an ischemic attack. Our laboratory demonstrated that its protection against myocardial dysfunction occurs by the low molecular weight (LMW) isoform of FGF2, while the high molecular weight (HMW) isoforms are associated with a worsening in post-ischemic recovery of cardiac function. LMW FGF2-mediated cardioprotection is facilitated by activation of multiple kinases, including PKCalpha, PKCepsilon, and ERK, and inhibition of p38 and JNK. OBJECTIVE Yet, the substrates of those kinases associated with LMW FGF2-induced cardioprotection against myocardial dysfunction remain to be elucidated. METHODS AND RESULTS To identify substrates in LMW FGF2 improvement of post-ischemic cardiac function, mouse hearts expressing only LMW FGF2 were subjected to ischemia-reperfusion (I/R) injury and analyzed by a mass spectrometry (MS)-based quantitative phosphoproteomic strategy. MS analysis identified 50 phosphorylation sites from 7 sarcoendoplasmic reticulum (SR) proteins that were significantly altered in I/R-treated hearts only expressing LMW FGF2 compared to those hearts lacking FGF2. One of those phosphorylated SR proteins identified was phospholamban (PLB), which exhibited rapid, increased phosphorylation at Threonine-17 (Thr17) after I/R in hearts expressing only LMW FGF2; this was further validated using Selected Reaction Monitoring-based MS workflow. To demonstrate a mechanistic role of phospho-Thr17 PLB in LMW FGF2-mediated cardioprotection, hearts only expressing LMW FGF2 and those expressing only LMW FGF2 with a mutant PLB lacking phosphorylatable Thr17 (Thr17Ala PLB) were subjected to I/R. Hearts only expressing LMW FGF2 showed significantly improved recovery of cardiac function following I/R (p < 0.05), and this functional improvement was significantly abrogated in hearts expressing LMW FGF2 and Thr17Ala PLB (p < 0.05). CONCLUSION The findings indicate that LMW FGF2 modulates intracellular calcium handling/cycling via regulatory changes in SR proteins essential for recovery from I/R injury, and thereby protects the heart from post-ischemic cardiac dysfunction.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Aruna B Wijeratne
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Brian B Oloizia
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Yu Zhang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Jo El J Schultz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America.
| |
Collapse
|
25
|
Dopamine receptor D3 agonist (Pramipexole) reduces morphine-induced cardiac fibrosis. Biochem Biophys Res Commun 2020; 529:1080-1085. [PMID: 32819568 DOI: 10.1016/j.bbrc.2020.06.137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 01/30/2023]
Abstract
Morphine is routinely used for pain management in heart failure patients. However, extended morphine exposure associates with major adverse cardiovascular events. Reports link the dopamine receptor D2-family with morphine-induced nociception modulation. This study first assessed whether morphine induces cardiac remodeling in healthy mice, then whether DRD3 agonist (DRD3ag, D2-family member) adjunct therapy prevents morphine-induced cardiac remodeling. Mice received morphine (2 mg/kg/day i. p.) for 7 days (D7) and were either euthanized at D7 or kept 7 more days without morphine (i.e. withdrawal period, D8-D14): G1, morphine; G2, morphine/DRD3ag; G3, morphine + withdrawal; G4, morphine/DRD3ag + withdrawal; G5, morphine + withdrawal/DRD3ag. A separate cohort of animals were used as naïve tissues. We evaluated functional and molecular parameters of cardiac remodeling. Although we did not observe significant differences in systolic function, morphine induced both interstitial fibrosis and cardiomyocyte hypertrophy. Interestingly, DRD3ag abolished these effects. Compared to naïve tissues, collagen 1 increased after withdrawal in G3 and G4 and collagen 3 increased in G1-G4 but at higher levels in G1 and G2. Only G5 did not show collagen differences compared to naïve, suggesting DRD3ag treatment during withdrawal may be beneficial and prevent morphine-induced fibrosis. Smad2/3 phosphorylation increased during withdrawal, indicating a likely upstream pathway for the observed morphine-induced fibrosis. Overall, our data suggest that DRD3ag adjunct therapy decreases morphine-induced adverse cardiac remodeling.
Collapse
|
26
|
Ostadal B, Ostadalova I, Szarszoi O, Netuka I, Olejnickova V, Hlavackova M. Sex-dependent effect of perinatal hypoxia on cardiac tolerance to oxygen deprivation in adults. Can J Physiol Pharmacol 2020; 99:1-8. [PMID: 32687731 DOI: 10.1139/cjpp-2020-0310] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epidemiological studies have demonstrated a relationship between the adverse influence of perinatal development and increased risk of ischemic heart disease in adults. From negative factors to which the fetus is subjected, the most important is hypoxia. The fetus may experience hypoxic stress under different conditions, including pregnancy at high altitude, pregnancy with anemia, placental insufficiency, and heart, lung, and kidney disease. One of the most common insults during the early stages of postnatal development is hypoxemia due to congenital cyanotic heart defects. Experimental studies have demonstrated a link between early hypoxia and increased risk of ischemia/reperfusion injury (I/R) in adults. Furthermore, it has been observed that late myocardial effects of chronic hypoxia, experienced in early life, may be sex-dependent. Unlike in males, perinatal hypoxia significantly increased cardiac tolerance to acute I/R injury in adult females, expressed as decreased infarct size and lower incidence of ischemic arrhythmias. It was suggested that early hypoxia may result in sex-dependent programming of specific genes in the offspring with the consequence of increased cardiac susceptibility to I/R injury in adult males. These results would have important clinical implications, since cardiac sensitivity to oxygen deprivation in adult patients may be significantly influenced by perinatal hypoxia in a sex-dependent manner.
Collapse
Affiliation(s)
- B Ostadal
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - I Ostadalova
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - O Szarszoi
- Department of Cardiovascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - I Netuka
- Department of Cardiovascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - V Olejnickova
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.,Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - M Hlavackova
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
27
|
Li J, Sun D, Li Y. Novel Findings and Therapeutic Targets on Cardioprotection of Ischemia/ Reperfusion Injury in STEMI. Curr Pharm Des 2020; 25:3726-3739. [PMID: 31692431 DOI: 10.2174/1381612825666191105103417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Acute ST-segment elevation myocardial infarction (STEMI) remains a leading cause of morbidity and mortality around the world. A large number of STEMI patients after the infarction gradually develop heart failure due to the infarcted myocardium. Timely reperfusion is essential to salvage ischemic myocardium from the infarction, but the restoration of coronary blood flow in the infarct-related artery itself induces myocardial injury and cardiomyocyte death, known as ischemia/reperfusion injury (IRI). The factors contributing to IRI in STEMI are complex, and microvascular obstruction, inflammation, release of reactive oxygen species, myocardial stunning, and activation of myocardial cell death are involved. Therefore, additional cardioprotection is required to prevent the heart from IRI. Although many mechanical conditioning procedures and pharmacological agents have been identified as effective cardioprotective approaches in animal studies, their translation into the clinical practice has been relatively disappointing due to a variety of reasons. With new emerging data on cardioprotection in STEMI over the past few years, it is mandatory to reevaluate the effectiveness of "old" cardioprotective interventions and highlight the novel therapeutic targets and new treatment strategies of cardioprotection.
Collapse
Affiliation(s)
- Jianqiang Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Danghui Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| |
Collapse
|
28
|
Cadeddu Dessalvi C, Pepe A, Penna C, Gimelli A, Madonna R, Mele D, Monte I, Novo G, Nugara C, Zito C, Moslehi JJ, de Boer RA, Lyon AR, Tocchetti CG, Mercuro G. Sex differences in anthracycline-induced cardiotoxicity: the benefits of estrogens. Heart Fail Rev 2020; 24:915-925. [PMID: 31256318 DOI: 10.1007/s10741-019-09820-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Anthracyclines are the cornerstone for many oncologic treatments, but their cardiotoxicity has been recognized for several decades. Female subjects, especially before puberty and adolescence, or after menopause, seem to be more at increased risk, with the prognostic impact of this sex issue being less consistent compared to other cardiovascular risk factors. Several studies imply that sex differences could depend on the lack of the protective effect of sex hormones against the anthracycline-initiated damage in cardiac cells, or on differential mitochondria-related oxidative gene expression. This is also reflected by the results obtained with different diagnostic methods, such as cardiovascular biomarkers and imaging techniques (echocardiography, magnetic resonance, and nuclear medicine) in the diagnosis and monitoring of cardiotoxicity, confirming that sex differences exist. The same is true about protective strategies from anthracycline cardiotoxicity. Indeed, first studied to withstand oxidative damage in response to ischemia/reperfusion (I/R) injury, cardioprotection has different outcomes in men and women. A number of studies assessed the differences in I/R response between male and female hearts, with oxidative stress and apoptosis being shared mechanisms between the I/R and anthracyclines heart damage. Sex hormones can modulate these mechanisms, thus confirming their importance in the pathophysiology in cardioprotection not only from the ischemia/reperfusion damage, but also from anthracyclines, fueling further cardio-oncologic research on the topic.
Collapse
Affiliation(s)
| | - Alessia Pepe
- Magnetic Resonance Imaging Unit, Fondazione G. Monasterio C.N.R.- Regione Toscana, Pisa, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Gimelli
- Nuclear Medicine Unit, Fondazione G. Monasterio C.N.R.- Regione Toscana, Pisa, Italy
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy
| | - Donato Mele
- Cardiology Unit, Emergency Department, University Hospital of Ferrara, Ferrara, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities- Cardiology, University of Catania, Catania, Italy
| | - Giuseppina Novo
- Department of Cardiology, University of Palermo, Palermo, Italy
| | - Cinzia Nugara
- Department of Cardiology, University of Palermo, Palermo, Italy
| | - Concetta Zito
- Department of Clinical and Experimental Medicine - Cardiology, University of Messina, Messina, Italy
| | - Javid J Moslehi
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rudolf A de Boer
- University Medical Center Groningen, Department of Cardiology, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | | | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy. .,Interdepartmental Center for Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy.
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
29
|
Xu S, Xie F, Tian L, Fallah S, Babaei F, Manno SHC, Manno FAM, Zhu L, Wong KF, Liang Y, Ramalingam R, Sun L, Wang X, Plumb R, Gethings L, Lam YW, Cheng SH. Estrogen accelerates heart regeneration by promoting the inflammatory response in zebrafish. J Endocrinol 2020; 245:39-51. [PMID: 31977314 PMCID: PMC7040496 DOI: 10.1530/joe-19-0413] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022]
Abstract
Sexual differences have been observed in the onset and prognosis of human cardiovascular diseases, but the underlying mechanisms are not clear. Here, we found that zebrafish heart regeneration is faster in females, can be accelerated by estrogen and is suppressed by the estrogen-antagonist tamoxifen. Injuries to the zebrafish heart, but not other tissues, increased plasma estrogen levels and the expression of estrogen receptors, especially esr2a. The resulting endocrine disruption induces the expression of the female-specific protein vitellogenin in male zebrafish. Transcriptomic analyses suggested heart injuries triggered pronounced immune and inflammatory responses in females. These responses, previously shown to elicit heart regeneration, could be enhanced by estrogen treatment in males and reduced by tamoxifen in females. Furthermore, a prior exposure to estrogen preconditioned the zebrafish heart for an accelerated regeneration. Altogether, this study reveals that heart regeneration is modulated by an estrogen-inducible inflammatory response to cardiac injury. These findings elucidate a previously unknown layer of control in zebrafish heart regeneration and provide a new model system for the study of sexual differences in human cardiac repair.
Collapse
Affiliation(s)
- Shisan Xu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Fangjing Xie
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Li Tian
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Samane Fallah
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Fatemeh Babaei
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Sinai H C Manno
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Francis A M Manno
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, New South Wales, Australia
| | - Lina Zhu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Kin Fung Wong
- Department of Biomedical Engineering, Polytechnic University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Yimin Liang
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Rajkumar Ramalingam
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Lei Sun
- Department of Biomedical Engineering, Polytechnic University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Xin Wang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Robert Plumb
- Waters Technologies Corporation, Milford, Massachusetts, USA
| | - Lee Gethings
- Waters Technologies Corporation, Milford, Massachusetts, USA
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
- Correspondence should be addressed to Y W Lam or S H Cheng: or
| | - Shuk Han Cheng
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
- State Key Laboratory of Marine Pollution (SKLMP) at City University of Hong Kong, Hong Kong SAR, People’s Republic of China
- Department of Materials Science and Engineering, College of Science and Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
- Correspondence should be addressed to Y W Lam or S H Cheng: or
| |
Collapse
|
30
|
Tower J, Pomatto LCD, Davies KJA. Sex differences in the response to oxidative and proteolytic stress. Redox Biol 2020; 31:101488. [PMID: 32201219 PMCID: PMC7212483 DOI: 10.1016/j.redox.2020.101488] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/16/2022] Open
Abstract
Sex differences in diseases involving oxidative and proteolytic stress are common, including greater ischemic heart disease, Parkinson disease and stroke in men, and greater Alzheimer disease in women. Sex differences are also observed in stress response of cells and tissues, where female cells are generally more resistant to heat and oxidative stress-induced cell death. Studies implicate beneficial effects of estrogen, as well as cell-autonomous effects including superior mitochondrial function and increased expression of stress response genes in female cells relative to male cells. The p53 and forkhead box (FOX)-family genes, heat shock proteins (HSPs), and the apoptosis and autophagy pathways appear particularly important in mediating sex differences in stress response.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA90089, USA.
| | - Laura C D Pomatto
- National Institute on General Medical Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kelvin J A Davies
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA90089, USA; Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, USA
| |
Collapse
|
31
|
Fetal Hypoxia Impacts on Proliferation and Differentiation of Sca-1 + Cardiac Progenitor Cells and Maturation of Cardiomyocytes: A Role of MicroRNA-210. Genes (Basel) 2020; 11:genes11030328. [PMID: 32244901 PMCID: PMC7140790 DOI: 10.3390/genes11030328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is one of the most frequent and severe stresses to an organism’s homeostatic mechanisms, and hypoxia during gestation has profound adverse effects on the heart development increasing the occurrence of congenital heart defects (CHDs). Cardiac progenitor cells (CPCs) are responsible for early heart development and the later occurrence of heart disease. However, the mechanism of how hypoxic stress affects CPC fate decisions and contributes to CHDs remains a topic of debate. Here we examined the effect of hypoxic stress on the regulations of CPC fate decisions and the potential mechanism. We found that experimental induction of hypoxic responses compromised CPC function by regulating CPC proliferation and differentiation and restraining cardiomyocyte maturation. In addition, echocardiography indicated that fetal hypoxia reduced interventricular septum thickness at diastole and the ejection time, but increased the heart rate, in mouse young adult offspring with a gender-related difference. Further study revealed that hypoxia upregulated microRNA-210 expression in Sca-1+ CPCs and impeded the cell differentiation. Blockage of microRNA-210 with LNA-anti-microRNA-210 significantly promoted differentiation of Sca-1+ CPCs into cardiomyocytes. Thus, the present findings provide clear evidence that hypoxia alters CPC fate decisions and reveal a novel mechanism of microRNA-210 in the hypoxic effect, raising the possibility of microRNA-210 as a potential therapeutic target for heart disease.
Collapse
|
32
|
Bøtker HE, Cabrera-Fuentes HA, Ruiz-Meana M, Heusch G, Ovize M. Translational issues for mitoprotective agents as adjunct to reperfusion therapy in patients with ST-segment elevation myocardial infarction. J Cell Mol Med 2020; 24:2717-2729. [PMID: 31967733 PMCID: PMC7077531 DOI: 10.1111/jcmm.14953] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Pre‐clinical studies have indicated that mitoprotective drugs may add cardioprotection beyond rapid revascularization, antiplatelet therapy and risk modification. We review the clinical efficacy of mitoprotective drugs that have progressed to clinical testing comprising cyclosporine A, KAI‐9803, MTP131 and TRO 40303. Whereas cyclosporine may reduce infarct size in patients undergoing primary angioplasty as evaluated by release of myocardial ischaemic biomarkers and infarct size imaging, the other drugs were not capable of demonstrating this effect in the clinical setting. The absent effect leaves the role of the mitochondrial permeability transition pore for reperfusion injury in humans unanswered and indicates that targeting one single mechanism to provide mitoprotection may not be efficient. Moreover, the lack of effect may relate to favourable outcome with current optimal therapy, but conditions such as age, sex, diabetes, dyslipidaemia and concurrent medications may also alter mitochondrial function. However, as long as the molecular structure of the pore remains unknown and specific inhibitors of its opening are lacking, the mitochondrial permeability transition pore remains a target for alleviation of reperfusion injury. Nevertheless, taking conditions such as ageing, sex, comorbidities and co‐medication into account may be of paramount importance during the design of pre‐clinical and clinical studies testing mitoprotective drugs.
Collapse
Affiliation(s)
- Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Hector Alejandro Cabrera-Fuentes
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme and Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore.,Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, Mexico.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russian Federation
| | - Marisol Ruiz-Meana
- Vall d'Hebron Institut de Recerca, University Hospital Vall d'Hebron-Universitat Autònoma, Barcelona, Spain.,Centro de Investigación Biomédica en Red-CV, CIBER-CV, Spain
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen. Medical School, Essen, Germany
| | - Michel Ovize
- CarMeN Laboratory, Hôpital Louis Pradel, Hospices Civils de Lyon, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, INSERM U1060, Lyon, France
| |
Collapse
|
33
|
Changes in nitric oxide synthase levels are associated with impaired cardiac function and tolerance to ischemia-reperfusion injury in male rats with transient congenital hypothyroidism. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1103-1111. [PMID: 31940052 DOI: 10.1007/s00210-020-01812-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
Transient congenital hypothyroidism (TCH) has long-lasting consequences on the cardiovascular system during adulthood. The aim of this study was to determine whether nitric oxide (NO) and NO-producing enzymes are involved in impaired cardiac function as well as decreased tolerance to ischemia-reperfusion (IR) injury in adult male rats with TCH. Pregnant rats were divided into control and hypothyroid groups. Male offspring rats were categorized in control and hypothyroid (TCH) groups at week 16. Levels of NOx (nitrate+nitrite) and neuronal NOS (nNOS), inducible NOS (iNOS), and endothelial NOS (eNOS) were measured in hearts of rats and isolated perfused hearts from both groups were subjected to IR. Levels of NOx and NOSs were also measured in both groups after ischemia. Compared with controls, heart NOx levels were higher at baseline (48.0 ± 4.9 vs. 35.0 ± 2.6 μmol/L; P = 0.034) and following IR (103.6 ± 4.2 vs. 70.2 ± 2.7 μmol/L; P < 0.001) in rat with TCH. At baseline, compared with controls, heart iNOS and nNOS levels were significantly higher in rats with TCH (6.12 ± 0.34 vs. 4.78 ± 0.27 ng/mg protein; P = 0.008 for iNOS and 4.87 ± 0.28 vs. 3.55 ± 0.23 ng/mg protein; P = 0.003 for nNOS). Following IR, in rats with TCH, heart iNOS levels increased (11.75 ± 2.02 vs. 6.12 ± 0.34, ng/mg protein; P = 0.015) whereas nNOS level decreased (4.10 ± 0.25 vs. 4.87 ± 0.28 ng/mg protein; P = 0.063). Adverse effects of TCH on cardiac function are associated with increased ratio of iNOS/eNOS; in addition, increased heart nNOS levels are involved in impaired cardiac function while its decrease is associated with decreased tolerance to IR injury.
Collapse
|
34
|
Ghimire A, Bisset ES, Howlett SE. Ischemia and reperfusion injury following cardioplegic arrest is attenuated by age and testosterone deficiency in male but not female mice. Biol Sex Differ 2019; 10:42. [PMID: 31443710 PMCID: PMC6708213 DOI: 10.1186/s13293-019-0256-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular disease increases with age in both sexes. Treatment can require cardiac surgery, where the hearts are pre-treated with protective cardioplegic solution before ischemia and reperfusion (I/R). While endogenous estrogen is beneficial in I/R, whether testosterone is involved is uncertain and whether age modifies responses to I/R is unclear. We investigated sex- and age-specific differences in I/R injury in the hearts pre-treated with clinically relevant cardioplegic solution. METHODS The hearts were isolated from young (6-9 months) and old (20-28 months) mice of both sexes and perfused (Langendorff) with Krebs-Henseleit buffer (15 min, 37 °C), followed by St. Thomas' two cardioplegia (6 min, 6-7 °C), global ischemia (90 min, 23-24 °C), and reperfusion (30 min, 37 °C). The hearts were perfused with triphenyltetrazolium chloride to quantify infarct area. Testosterone's role was investigated in gonadectomized (GDX, 6-9 months) male mice; serum testosterone and estradiol were measured with ELISA assays. RESULTS Left ventricular developed pressure (LVDP) recovered to 67.3 ± 7.4% in the old compared to 21.8 ± 9.2% in the young male hearts (p < 0.05). Similar results were seen for rates of pressure development (+dP/dt) and decay (-dP/dt). Infarct areas were smaller in the old male hearts (16.6 ± 1.6%) than in the younger hearts (55.8 ± 1.2%, p < 0.05). By contrast, the hearts from young and old females exhibited a similar post-ischemic functional recovery and no age-dependent difference in infarcts. There was a sex difference in the young group, where ventricular function (LVDP, +dP/dt, -dP/dt) recovered better and infarcts were smaller in females than males. Estradiol levels were highest in young females. Testosterone was high in young males but low in females and old males, which suggested beneficial effects of low testosterone. Indeed, the hearts from GDX males exhibited much better recovery of LVDP in reperfusion than that from intact males (values were 64.4 ± 7.5 % vs. 21.8 ± 9.2%; p < 0.05). The GDX hearts also had smaller infarcts than the hearts from intact males (p < 0.05). CONCLUSIONS Although age had no effect on susceptibility to I/R injury after cardioplegic arrest in females, it actually protected against injury in older males. Our findings indicate that low testosterone may be protective against I/R injury following cardioplegic arrest in older males.
Collapse
Affiliation(s)
- Anjali Ghimire
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, PO Box 15000, Halifax, Nova Scotia B3H 4R2 Canada
| | - Elise S. Bisset
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, PO Box 15000, Halifax, Nova Scotia B3H 4R2 Canada
| | - Susan E. Howlett
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, PO Box 15000, Halifax, Nova Scotia B3H 4R2 Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia Canada
| |
Collapse
|
35
|
Atrioventricular mechanical coupling and major adverse cardiac events in female patients following acute ST elevation myocardial infarction. Int J Cardiol 2019; 299:31-36. [PMID: 31300172 DOI: 10.1016/j.ijcard.2019.06.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND Sex-specific outcome data following myocardial infarction (MI) are inconclusive with some evidence suggesting association of female sex and increased major adverse cardiac events (MACE). Since mechanistic principles remain elusive, we aimed to quantify the underlying phenotype using cardiovascular magnetic resonance (CMR) quantitative deformation imaging and tissue characterisation. METHODS In total, 795 ST-elevation MI patients underwent post-interventional CMR imaging. Feature-tracking (CMR-FT) was performed in a blinded core-laboratory. Left ventricular function was quantified using ejection fraction (LVEF) and global longitudinal/circumferential/radial strains (GLS/GCS/GRS). Left atrial function was assessed by reservoir (εs), conduit (εe) and booster-pump strains (εa). Tissue characterisation included infarct size, microvascular obstruction and area at risk. Primary endpoint was the occurrence of MACE within 1 year. RESULTS Female sex was associated with increased MACE (HR 1.96, 95% CI 1.13-3.42, p = 0.017) but not independently of baseline confounders (p = 0.526) with women being older, more often diabetic and hypertensive (p < 0.001) and of higher Killip-class (p = 0.010). Tissue characterisation was similar between sexes. Women showed impaired atrial (εs p = 0.011, εe p < 0.001) but increased systolic ventricular mechanics (GLS p = 0.001, LVEF p = 0.048). While atrial and ventricular function predicted MACE in men only LV GLS and GCS were associated with MACE in women irrespective of confounders (GLS p = 0.036, GCS p = 0.04). CONCLUSION In men ventricular systolic contractility is impaired and volume assessments precisely stratify elevated risks. In contrast, women experience reduced atrial but increased ventricular systolic strain. This may reflect ventricular diastolic failure with systolic compensation, which is independently associated with MACE adding incremental value to sex-specific prognosis evaluation.
Collapse
|
36
|
Ostadal B, Drahota Z, Houstek J, Milerova M, Ostadalova I, Hlavackova M, Kolar F. Developmental and sex differences in cardiac tolerance to ischemia-reperfusion injury: the role of mitochondria 1. Can J Physiol Pharmacol 2019; 97:808-814. [PMID: 30893574 DOI: 10.1139/cjpp-2019-0060] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Age and sex play an essential role in the cardiac tolerance to ischemia-reperfusion injury: cardiac resistance significantly decreases during postnatal maturation and the female heart is more tolerant than the male myocardium. It is widely accepted that mitochondrial dysfunction, and particularly mitochondrial permeability transition pore (MPTP) opening, plays a major role in determining the extent of cardiac ischemia-reperfusion injury. We have observed that the MPTP sensitivity to the calcium load differs in mitochondria isolated from neonatal and adult myocardium, as well as from adult male and female hearts. Neonatal and female mitochondria are more resistant both in the extent and in the rate of mitochondrial swelling induced by high calcium concentration. Our data further suggest that age- and sex-dependent specificity of the MPTP is not the result of different amounts of ATP synthase and cyclophilin D: neonatal and adult hearts, similarly as the male and female hearts, contain comparable amounts of MPTP and its regulatory protein cyclophilin D. We can speculate that the lower sensitivity of MPTP to the calcium-induced swelling may be related to the higher ischemic tolerance of both neonatal and female myocardium.
Collapse
Affiliation(s)
- B Ostadal
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - Z Drahota
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - J Houstek
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - M Milerova
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - I Ostadalova
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - M Hlavackova
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| | - F Kolar
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic.,Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague 4 Czech Republic
| |
Collapse
|
37
|
Fels JA, Manfredi G. Sex Differences in Ischemia/Reperfusion Injury: The Role of Mitochondrial Permeability Transition. Neurochem Res 2019; 44:2336-2345. [PMID: 30863968 DOI: 10.1007/s11064-019-02769-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/23/2022]
Abstract
Brain and heart ischemia are among the leading causes of death and disability in both men and women, but there are significant sex differences in the incidence and severity of these diseases. Ca2+ dysregulation in response to ischemia/reperfusion injury (I/RI) is a well-recognized pathogenic mechanism leading to the death of affected cells. Excess intracellular Ca2+ causes mitochondrial matrix Ca2+ overload that can result in mitochondrial permeability transition (MPT), which can have severe consequences for mitochondrial function and trigger cell death. Recent findings indicate that estrogens and their related receptors are involved in the regulation of MPT, suggesting that sex differences in I/RI could be linked to estrogen-dependent modulation of mitochondrial Ca2+. Here, we review the evidence supporting sex differences in I/RI and the role of estrogen and estrogen receptors in producing these differences, the involvement of mitochondrial Ca2+ overload in disease pathogenesis, and the estrogen-dependent modulation of MPT that may contribute to sex differences.
Collapse
Affiliation(s)
- Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st St., RR506, New York, NY, 10065, USA.,Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st St., RR506, New York, NY, 10065, USA.
| |
Collapse
|
38
|
Tian J, Wang X, Tian J, Yu B. Gender differences in plaque characteristics of nonculprit lesions in patients with coronary artery disease. BMC Cardiovasc Disord 2019; 19:45. [PMID: 30808307 PMCID: PMC6390304 DOI: 10.1186/s12872-019-1023-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/15/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Although numerous reports suggest sex-related differences in atherosclerosis, limited data describing gender-associated differences in plaque morphology and composition are currently available. The aim of the present study was to compare coronary nonculprit plaque characteristics in women and men with coronary artery disease (CAD) by optical coherence tomography (OCT). METHODS This was a retrospective study. A total of 187 nonculprit plaques were identified in 103 patients with CAD who underwent OCT imaging of all 3 coronary arteries. These patients included 77 (74.8%) men and 26 (25.2%) women. RESULTS Female patients were significantly older than males (mean age, 70.8 ± 7.3 vs 60.8 ± 9.8 years; P < 0.001) and less likely to be current smokers (P = 0.007). OCT analysis included the presence of lipid-rich plaque, maximum lipid arc, lipid-core length, lipid index (LI), fibrous cap thickness, and the incidence of thin-cap fibroatheroma (TCFA). Nonculprit plaques in men exhibited greater lipid-core length and LI compared with those of women (9.4 ± 4.5 vs. 7.3 ± 4.3 mm, P = 0.024; 1615.1 ± 893.8 vs. 1237.8 ± 859.8, P = 0.035, respectively). In the univariate linear regression model, sex and current smoker were all associated with a larger LI, whereas only use of statin was independent risk factor for a larger LI in multivariate analysis. CONCLUSIONS Coronary nonculprit plaques in male patients with CAD contain larger lipid cores than those of female patients.
Collapse
Affiliation(s)
- Jiangtian Tian
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xuedong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Bo Yu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China. .,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
39
|
Romiti GF, Cangemi R, Toriello F, Ruscio E, Sciomer S, Moscucci F, Vincenti M, Crescioli C, Proietti M, Basili S, Raparelli V. Sex-Specific Cut-Offs for High-Sensitivity Cardiac Troponin: Is Less More? Cardiovasc Ther 2019; 2019:9546931. [PMID: 31772621 PMCID: PMC6739766 DOI: 10.1155/2019/9546931] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Management of patients presenting to the Emergency Department with chest pain is continuously evolving. In the setting of acute coronary syndrome, the availability of high-sensitivity cardiac troponin assays (hs-cTn) has allowed for the development of algorithms aimed at rapidly assessing the risk of an ongoing myocardial infarction. However, concerns were raised about the massive application of such a simplified approach to heterogeneous real-world populations. As a result, there is a potential risk of underdiagnosis in several clusters of patients, including women, for whom a lower threshold for hs-cTn was suggested to be more appropriate. Implementation in clinical practice of sex-tailored cut-off values for hs-cTn represents a hot topic due to the need to reduce inequality and improve diagnostic performance in females. The aim of this review is to summarize current evidence on sex-specific cut-off values of hs-cTn and their application and usefulness in clinical practice. We also offer an extensive overview of thresholds reported in literature and of the mechanisms underlying such differences among sexes, suggesting possible explanations about debated issues.
Collapse
Affiliation(s)
- Giulio Francesco Romiti
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Roberto Cangemi
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Filippo Toriello
- Division of Cardiology, San Paolo Hospital, Department of Health Sciences, University of Milan, Milan, Italy
| | - Eleonora Ruscio
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Susanna Sciomer
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza–University of Rome, Rome, Italy
| | - Federica Moscucci
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza–University of Rome, Rome, Italy
| | - Marianna Vincenti
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Marco Proietti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Valeria Raparelli
- Department of Experimental Medicine, Sapienza–University of Rome, Rome, Italy
- Center for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
40
|
Puglisi R, Mattia G, Carè A, Marano G, Malorni W, Matarrese P. Non-genomic Effects of Estrogen on Cell Homeostasis and Remodeling With Special Focus on Cardiac Ischemia/Reperfusion Injury. Front Endocrinol (Lausanne) 2019; 10:733. [PMID: 31708877 PMCID: PMC6823206 DOI: 10.3389/fendo.2019.00733] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
This review takes into consideration the main mechanisms involved in cellular remodeling following an ischemic injury, with special focus on the possible role played by non-genomic estrogen effects. Sex differences have also been considered. In fact, cardiac ischemic events induce damage to different cellular components of the heart, such as cardiomyocytes, vascular cells, endothelial cells, and cardiac fibroblasts. The ability of the cardiovascular system to counteract an ischemic insult is orchestrated by these cell types and is carried out thanks to a number of complex molecular pathways, including genomic (slow) or non-genomic (fast) effects of estrogen. These pathways are probably responsible for differences observed between the two sexes. Literature suggests that male and female hearts, and, more in general, cardiovascular system cells, show significant differences in many parameters under both physiological and pathological conditions. In particular, many experimental studies dealing with sex differences in the cardiovascular system suggest a higher ability of females to respond to environmental insults in comparison with males. For instance, as cells from females are more effective in counteracting the ischemia/reperfusion injury if compared with males, a role for estrogen in this sex disparity has been hypothesized. However, the possible involvement of estrogen-dependent non-genomic effects on the cardiovascular system is still under debate. Further experimental studies, including sex-specific studies, are needed in order to shed further light on this matter.
Collapse
Affiliation(s)
- Rossella Puglisi
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Mattia
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Marano
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Walter Malorni
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
- School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Matarrese
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Paola Matarrese
| |
Collapse
|
41
|
Role of miRNA in the Regulatory Mechanisms of Estrogens in Cardiovascular Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6082387. [PMID: 30671171 PMCID: PMC6317101 DOI: 10.1155/2018/6082387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/13/2018] [Indexed: 12/24/2022]
Abstract
Cardiovascular diseases are a worldwide health problem and are the leading cause of mortality in developed countries. Together with experimental data, the lower incidence of cardiovascular diseases in women than in men of reproductive age points to the influence of sex hormones at the cardiovascular level and suggests that estrogens play a protective role against cardiovascular disease and that this role is also modified by ageing. Estrogens affect cardiovascular function via their specific estrogen receptors to trigger gene expression changes at the transcriptional level. In addition, emerging studies have proposed a role for microRNAs in the vascular effects mediated by estrogens. miRNAs regulate gene expression by repressing translational processes and have been estimated to be involved in the regulation of approximately 30% of all protein-coding genes in mammals. In this review, we highlight the current knowledge of the role of estrogen-sensitive miRNAs, and their influence in regulating vascular ageing.
Collapse
|
42
|
Kingma JG. Effect of Platelet GPIIb/IIIa Receptor Blockade With MK383 on Infarct Size and Myocardial Blood Flow in a Canine Reocclusion Model. J Cardiovasc Pharmacol Ther 2018; 24:182-192. [PMID: 30428694 DOI: 10.1177/1074248418808389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Platelet activation and aggregation during ischemia influence reperfusion-related myocyte necrosis, myocardial perfusion at the microvascular level, and thereby eventual recovery of cardiac performance. Inhibition of platelet activity therefore represents a worthwhile target to reduce cellular injury. The current study examined the effects of MK383 (tirofiban), a potent inhibitor of platelet aggregation, on infarct size and myocardial perfusion in canine subjects to either reocclusion (ie, 120-minute + 60-minute ischemia with intervening reperfusion) or prolonged occlusion (ie, 3 hours) followed by reperfusion (180 minutes). Platelet aggregation, infarct size (tetrazolium staining), coronary blood flow (flow probe), coronary vascular reserve, and myocardial perfusion (microspheres) were evaluated. MK383, administered at the time of reperfusion, produced a modest reduction of tissue necrosis (compared to saline-treated controls) in the reocclusion and prolonged occlusion studies. Blood flow in the infarct-related artery after coronary occlusion was comparable between treatment groups, as was myocardial perfusion in the deeper layers of the ischemic region; coronary vascular reserve decreased progressively during reperfusion. Of note, compensatory changes in blood flow within the adjacent nonischemic myocardium were not observed. In conclusion, we report that that limiting platelet aggregation during reperfusion impacted infarct development. Continued investigation into the mechanisms by which inhibition of platelet activity protects myocardium against ischemia-reperfusion injury and improves clinical outcomes is necessary.
Collapse
Affiliation(s)
- John G Kingma
- Department of Medicine, Faculty of Medicine, Laval University, Pavillon Ferdinand Vandry, Quebec, Canada
| |
Collapse
|
43
|
|
44
|
Randhawa PK, Bali A, Virdi JK, Jaggi AS. Conditioning-induced cardioprotection: Aging as a confounding factor. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:467-479. [PMID: 30181694 PMCID: PMC6115349 DOI: 10.4196/kjpp.2018.22.5.467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 05/15/2018] [Indexed: 01/15/2023]
Abstract
The aging process induces a plethora of changes in the body including alterations in hormonal regulation and metabolism in various organs including the heart. Aging is associated with marked increase in the vulnerability of the heart to ischemia-reperfusion injury. Furthermore, it significantly hampers the development of adaptive response to various forms of conditioning stimuli (pre/post/remote conditioning). Aging significantly impairs the activation of signaling pathways that mediate preconditioning-induced cardioprotection. It possibly impairs the uptake and release of adenosine, decreases the number of adenosine transporter sites and down-regulates the transcription of adenosine receptors in the myocardium to attenuate adenosine-mediated cardioprotection. Furthermore, aging decreases the expression of peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α) and subsequent transcription of catalase enzyme which subsequently increases the oxidative stress and decreases the responsiveness to preconditioning stimuli in the senescent diabetic hearts. In addition, in the aged rat hearts, the conditioning stimulus fails to phosphorylate Akt kinase that is required for mediating cardioprotective signaling in the heart. Moreover, aging increases the concentration of Na+ and K+, connexin expression and caveolin abundance in the myocardium and increases the susceptibility to ischemia-reperfusion injury. In addition, aging also reduces the responsiveness to conditioning stimuli possibly due to reduced kinase signaling and reduced STAT-3 phosphorylation. However, aging is associated with an increase in MKP-1 phosphorylation, which dephosphorylates (deactivates) mitogen activated protein kinase that is involved in cardioprotective signaling. The present review describes aging as one of the major confounding factors in attenuating remote ischemic preconditioning-induced cardioprotection along with the possible mechanisms.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | - Anjana Bali
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur 148002, India
| | - Jasleen Kaur Virdi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| |
Collapse
|
45
|
Bøtker HE, Hausenloy D, Andreadou I, Antonucci S, Boengler K, Davidson SM, Deshwal S, Devaux Y, Di Lisa F, Di Sante M, Efentakis P, Femminò S, García-Dorado D, Giricz Z, Ibanez B, Iliodromitis E, Kaludercic N, Kleinbongard P, Neuhäuser M, Ovize M, Pagliaro P, Rahbek-Schmidt M, Ruiz-Meana M, Schlüter KD, Schulz R, Skyschally A, Wilder C, Yellon DM, Ferdinandy P, Heusch G. Practical guidelines for rigor and reproducibility in preclinical and clinical studies on cardioprotection. Basic Res Cardiol 2018; 113:39. [PMID: 30120595 PMCID: PMC6105267 DOI: 10.1007/s00395-018-0696-8] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/18/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle-Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Derek Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- The National Institute of Health Research, University College London Hospitals Biomedial Research Centre, Research and Development, London, UK
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
- Yon Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Salvatore Antonucci
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Kerstin Boengler
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Soni Deshwal
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Di Lisa
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Moises Di Sante
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - David García-Dorado
- Experimental Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), IIS-Fundación Jiménez Díaz, CIBERCV, Madrid, Spain
| | - Efstathios Iliodromitis
- Second Department of Cardiology, Faculty of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nina Kaludercic
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Via Ugo Bassi 58/B, 35121, Padua, Italy
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Markus Neuhäuser
- Department of Mathematics and Technology, Koblenz University of Applied Science, Remagen, Germany
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, Essen, Germany
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France
- UMR, 1060 (CarMeN), Université Claude Bernard, Lyon1, Villeurbanne, France
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Michael Rahbek-Schmidt
- Department of Cardiology, Aarhus University Hospital, Palle-Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Marisol Ruiz-Meana
- Experimental Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Catherine Wilder
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
46
|
Ramratnam M, Kenny B, Kyle JW, Wiedmeyer B, Hacker TA, Barefield DY, McNally EM, Makielski JC. Transgenic overexpression of the SUR2A-55 splice variant in mouse heart reduces infract size and promotes protective mitochondrial function. Heliyon 2018; 4:e00677. [PMID: 29998196 PMCID: PMC6037880 DOI: 10.1016/j.heliyon.2018.e00677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/28/2018] [Indexed: 01/21/2023] Open
Abstract
ATP-sensitive potassium channels found in both the sarcolemma (sarcKATP) and mitochondria (mitoKATP) of cardiomyocytes are important mediators of cardioprotection during ischemic heart disease. Sulfonylurea receptor isoforms (SUR2), encoded by Abcc9, an ATP-binding cassette family member, form regulatory subunits of the sarcKATP channel and are also thought to regulate mitoKATP channel activity. A short-form splice variant of SUR2 (SUR2A-55) was previously shown to target mitochondria and display diaxoxide and ATP insensitive KATP activity when co-expressed with the inward rectifier channels Kir6.2 and Kir6.1. We hypothesized that mice with cardiac specific overexpression of SUR2A-55 would mediate cardioprotection from ischemia by altering mitoKATP properties. Mice overexpressing SUR2A-55 (TGSUR2A-55) in cardiomyocytes were generated and showed no significant difference in echocardiographic measured chamber dimension, percent fractional shortening, heart to body weight ratio, or gross histologic features compared to normal mice at 11–14 weeks of age. TGSUR2A-55 had improved hemodynamic functional recovery and smaller infarct size after ischemia reperfusion injury compared to WT mice in an isolated hanging heart model. The mitochondrial membrane potential of TGSUR2A-55 mice was less sensitive to ATP, diazoxide, and Ca2+ loading. These data suggest that the SUR2A-55 splice variant favorably affects mitochondrial function leading to cardioprotection. These data support a role for the regulation of mitoKATP activity by SUR2A-55.
Collapse
Affiliation(s)
- Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Barrett Kenny
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - John W Kyle
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Brandi Wiedmeyer
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Timothy A Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States
| | - Jonathan C Makielski
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
47
|
Burstein SR, Kim HJ, Fels JA, Qian L, Zhang S, Zhou P, Starkov AA, Iadecola C, Manfredi G. Estrogen receptor beta modulates permeability transition in brain mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2018; 1859:423-433. [PMID: 29550215 PMCID: PMC5912174 DOI: 10.1016/j.bbabio.2018.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 12/24/2022]
Abstract
Recent evidence highlights a role for sex and hormonal status in regulating cellular responses to ischemic brain injury and neurodegeneration. A key pathological event in ischemic brain injury is the opening of a mitochondrial permeability transition pore (MPT) induced by excitotoxic calcium levels, which can trigger irreversible damage to mitochondria accompanied by the release of pro-apoptotic factors. However, sex differences in brain MPT modulation have not yet been explored. Here, we show that mitochondria isolated from female mouse forebrain have a lower calcium threshold for MPT than male mitochondria, and that this sex difference depends on the MPT regulator cyclophilin D (CypD). We also demonstrate that an estrogen receptor beta (ERβ) antagonist inhibits MPT and knockout of ERβ decreases the sensitivity of mitochondria to the CypD inhibitor, cyclosporine A. These results suggest a functional relationship between ERβ and CypD in modulating brain MPT. Moreover, co-immunoprecipitation studies identify several ERβ binding partners in mitochondria. Among these, we investigate the mitochondrial ATPase as a putative site of MPT regulation by ERβ. We find that previously described interaction between the oligomycin sensitivity-conferring subunit of ATPase (OSCP) and CypD is decreased by ERβ knockout, suggesting that ERβ modulates MPT by regulating CypD interaction with OSCP. Functionally, in primary neurons and hippocampal slice cultures, modulation of ERβ has protective effects against glutamate toxicity and oxygen glucose deprivation, respectively. Taken together, these results reveal a novel pathway of brain MPT regulation by ERβ that could contribute to sex differences in ischemic brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Suzanne R Burstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Hyun Jeong Kim
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Liping Qian
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Sheng Zhang
- Proteomics and Mass Spectrometry Facility, 139 Biotechnology Building, Cornell University, 526 Campus Road, Ithaca, NY 14853, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Anatoly A Starkov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| |
Collapse
|
48
|
Yang GZ, Xue FS, Liu YY, Li HX, Liu Q, Liao X. Effects of enteral different-dose levothyroxinesodium pretreatment on serum thyroid hormone levels and myocardial ischemia-reperfusion injury. Perfusion 2018; 33:584-592. [PMID: 29722287 DOI: 10.1177/0267659118769228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The available evidence shows that perioperative oral thyroid hormone can significantly attenuate the postoperative decline in the serum hormone level and improve postoperative hemodynamic and prognostic parameters. However, there has been no study assessing the effects of preoperative oral different-dose thyroid hormone on serum hormone levels and myocardial ischemia-reperfusion injury (IRI) after cardiac surgery. METHODS Forty-eight healthy Wistar rats, aged 35 days, were randomly allocated into six groups: Group BC, Group C and four pretreatment groups in which the rats were given levothyroxine-sodium of 10 μg, 20 μg, 40 μg and 80 μg/100 g. On the eighth day, the serum thyroid hormone levels were determined and then an isolated heart ischemia-reperfusion model was established with a Langendorff apparatus. RESULTS Compared with Groups BC and C, serum thyroid hormone levels on the eighth day did not significantly change in Group 10 μg, but were significantly increased in Groups 20 μg, 40 μg and 80 μg. The cardiac enzyme myocardial-bound creatine kinase levels in the coronary effluent during reperfusion were significantly lower in Groups 10 μg and 20 μg and 40 μg than in Group C. The recovery rates of + dp/dtmax and - dp/dtmax at 30 min during reperfusion were significantly lower in Groups 40 μg and 80 μg than in Groups 10 μg and 20 μg. Compared with Group C, myocardial expressions of heat shock protein 70 and myosin heavy chain α were increased in the four experiment groups and myocardial expression of thyroid hormone receptor α1 was significantly increased in Groups 20 μg, 40 μg and 80 μg. CONCLUSIONS The pretreatment with enterally smaller doses levothyroxine-sodium does not significantly affect serum thyroid hormone levels and produces protection against myocardial IRI, whereas pretreatment with enterally larger doses of levothyroxine-sodium can only provide an attenuated or insignificant cardioprotection because of hyperthyroxinemia. Cardioprotection by levothyroxine-sodium pretreatment is probably attributable to increased myocardial expression of heat shock protein 70 and myosin heavy chain α.
Collapse
Affiliation(s)
- Gui-Zhen Yang
- 1 Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fu-Shan Xue
- 2 Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ya-Yang Liu
- 1 Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Xian Li
- 1 Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Liu
- 1 Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Liao
- 1 Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
49
|
Kingma JG. Inhibition of Na +/H + Exchanger With EMD 87580 does not Confer Greater Cardioprotection Beyond Preconditioning on Ischemia-Reperfusion Injury in Normal Dogs. J Cardiovasc Pharmacol Ther 2018; 23:254-269. [PMID: 29562750 DOI: 10.1177/1074248418755120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Postischemic accumulation of intracellular Na+ promotes calcium overload and contributes to cellular necrosis. Cardioprotection afforded by pharmacologic blockade of the sodium-hydrogen exchanger subtype 1 (NHE1) is thought to be more remarkable than that obtained by ischemic conditioning (IC). The window of protection provided by IC pretreatment is maintained even when performed up to 48 hours before ischemia. In addition, the perception exists that combined NHE1 inhibition plus IC produces greater than additive protection against ischemic injury. The current study compared the efficacy of NHE1 blockade by N-[2-methyl-4,5-bis(methylsulfonyl)-benzoyl]-guanidine (EMD 87580 5 mg/kg) combined with first- or second-window IC on ischemic tolerance in dogs subject to 90-minute acute ischemia and 180-minute reperfusion. Infarct size (tetrazolium staining), vascular responses, and myocardial perfusion (microspheres) were assessed. EMD 87580 given before ischemia or before reperfusion did not reduce infarct size (compared to vehicle-treated group). Significant protection against tissue necrosis was obtained by both first- and second-window IC, but additive cardioprotection (ie, greater than that afforded by IC) was not observed by treatment with EMD 87580. Vascular reactivity in the infarct-related artery was not preserved after ischemia-reperfusion in any of the experimental groups. Likewise, either the pharmacologic or the nonpharmacologic interventions did not modify myocardial perfusion. These data demonstrate that EMD 87580 did not protect against ischemia-reperfusion injury regardless of the time of drug administration. Combined EMD 87580 and IC did not antagonize protection that was achieved by either first- or second-window IC alone; no additive protection beyond preconditioning was obtained. Further study is necessary to assess the value of NHE1 blockers as protective agents against myocardial injury.
Collapse
Affiliation(s)
- J G Kingma
- 1 Faculty of Medicine, Department of Medicine, Laval University, Québec City, Québec, Canada
| |
Collapse
|
50
|
Pomatto LCD, Tower J, Davies KJA. Sexual Dimorphism and Aging Differentially Regulate Adaptive Homeostasis. J Gerontol A Biol Sci Med Sci 2018; 73:141-149. [PMID: 28525535 PMCID: PMC5861879 DOI: 10.1093/gerona/glx083] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/26/2017] [Indexed: 11/13/2022] Open
Abstract
External and internal stimuli cause modifications to gene and biochemical pathways. In turn, demonstrating that biological systems continuously make short-term adaptations both to set-points, and to the range of "normal" capacity, due to mild conditional changes, or to subtoxic, nondamaging levels of chemical agents. This is termed as "Adaptive Homeostasis," defined with the following: "The transient expansion or contraction of the homeostatic range in response to exposure to sub-toxic, nondamaging, signaling molecules or events, or the removal or cessation of such molecules or events." Research from several laboratories, including our own, found that adaptive homeostasis declines with age in organisms as diverse as worms, flies, and mammals, and decreases with senescence in mammalian cell cultures. We suggest that diminishing adaptive homeostasis may play a causal role as a factor responsible for the aging phenotype. Furthermore, although studies of humans, animals, and model organisms are often limited to a single sex, and cell culture studies may even be conducted with lines whose donor's sex was unknown, studies reveal distinct sexual dimorphism in adaptive homeostasis. Interestingly, although young males and females may exhibit dramatic differences in adaptive capacities and/or preferences, these distinctions are lost with age as adaptive homeostasis patterns converge.
Collapse
Affiliation(s)
- Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center
| | - John Tower
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California
| |
Collapse
|