1
|
Samani SL, Barlow SC, Freeburg LA, Jones TL, Poole M, Sarzynski MA, Zile MR, Shazly T, Spinale FG. Left ventricle function and post-transcriptional events with exercise training in pigs. PLoS One 2024; 19:e0292243. [PMID: 38306359 PMCID: PMC10836705 DOI: 10.1371/journal.pone.0292243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 09/14/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Standardized exercise protocols have been shown to improve overall cardiovascular fitness, but direct effects on left ventricular (LV) function, particularly diastolic function and relation to post-transcriptional molecular pathways (microRNAs (miRs)) are poorly understood. This project tested the central hypothesis that adaptive LV remodeling resulting from a large animal exercise training protocol, would be directly associated with specific miRs responsible for regulating pathways relevant to LV myocardial stiffness and geometry. METHODS AND RESULTS Pigs (n = 9; 25 Kg) underwent a 4 week exercise training protocol (10 degrees elevation, 2.5 mph, 10 min, 5 days/week) whereby LV chamber stiffness (KC) and regional myocardial stiffness (rKm) were measured by Doppler/speckle tracking echocardiography. Age and weight matched non-exercise pigs (n = 6) served as controls. LV KC fell by approximately 50% and rKm by 30% following exercise (both p < 0.05). Using an 84 miR array, 34 (40%) miRs changed with exercise, whereby 8 of the changed miRs (miR-19a, miR-22, miR-30e, miR-99a, miR-142, miR-144, miR-199a, and miR-497) were correlated to the change in KC (r ≥ 0.5 p < 0.05) and mapped to matrix and calcium handling processes. Additionally, miR-22 and miR-30e decreased with exercise and mapped to a localized inflammatory process, the inflammasome (NLRP-3, whereby a 2-fold decrease in NLRP-3 mRNA occurred with exercise (p < 0.05). CONCLUSION Chronic exercise reduced LV chamber and myocardial stiffness and was correlated to miRs that map to myocardial relaxation processes as well as local inflammatory pathways. These unique findings set the stage for utilization of myocardial miR profiling to identify underlying mechanisms by which exercise causes changes in LV myocardial structure and function.
Collapse
Affiliation(s)
- Stephanie L. Samani
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
| | - Shayne C. Barlow
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Lisa A. Freeburg
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
| | - Traci L. Jones
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Marlee Poole
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
| | - Mark A. Sarzynski
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States of America
| | - Michael R. Zile
- Division of Cardiology, RHJ Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC, United States of America
| | - Tarek Shazly
- College of Engineering and Computing, University of South Carolina, Columbia, SC, United States of America
| | - Francis G. Spinale
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States of America
- Columbia VA Health Care System, Columbia, SC, United States of America
- College of Engineering and Computing, University of South Carolina, Columbia, SC, United States of America
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States of America
| |
Collapse
|
2
|
Fisher SM, Murally AR, Rajabally Z, Almas T, Azhar M, Cheema FH, Malone A, Hasan B, Aslam N, Saidi J, O'Neill J, Hameed A. Large animal models to study effectiveness of therapy devices in the treatment of heart failure with preserved ejection fraction (HFpEF). Heart Fail Rev 2024; 29:257-276. [PMID: 37999821 DOI: 10.1007/s10741-023-10371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Our understanding of the complex pathophysiology of Heart failure with preserved ejection fraction (HFpEF) is limited by the lack of a robust in vivo model. Existing in-vivo models attempt to reproduce the four main phenotypes of HFpEF; ageing, obesity, diabetes mellitus and hypertension. To date, there is no in vivo model that represents all the haemodynamic characteristics of HFpEF, and only a few have proven to be reliable for the preclinical evaluation of potentially new therapeutic targets. HFpEF accounts for 50% of all the heart failure cases and its incidence is on the rise, posing a huge economic burden on the health system. Patients with HFpEF have limited therapeutic options available. The inadequate effectiveness of current pharmaceutical therapeutics for HFpEF has prompted the development of device-based treatments that target the hemodynamic changes to reduce the symptoms of HFpEF. However, despite the potential of device-based solutions to treat HFpEF, most of these therapies are still in the developmental stage and a relevant HFpEF in vivo model will surely expedite their development process. This review article outlines the major limitations of the current large in-vivo models in use while discussing how these designs have helped in the development of therapy devices for the treatment of HFpEF.
Collapse
Affiliation(s)
- Shane Michael Fisher
- Health Sciences Centre, UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Dublin, Ireland
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Anjali Rosanna Murally
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
- School of Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Zahra Rajabally
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
- School of Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Talal Almas
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, 123 St. Stephen's Green, Dublin, D02 YN77, Ireland
| | - Faisal H Cheema
- Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX, USA
| | - Andrew Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Babar Hasan
- Division of Cardiothoracic Sciences, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Nadeem Aslam
- Division of Cardiothoracic Sciences, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Jemil Saidi
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Jim O'Neill
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland.
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland.
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
3
|
Qin X, Cai P, Liu C, Chen K, Jiang X, Chen W, Li J, Jiao X, Guo E, Yu Y, Sun L, Tian H. Cardioprotective effect of ultrasound-targeted destruction of Sirt3-loaded cationic microbubbles in a large animal model of pathological cardiac hypertrophy. Acta Biomater 2023; 164:604-625. [PMID: 37080445 DOI: 10.1016/j.actbio.2023.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/22/2023]
Abstract
Pathological cardiac hypertrophy occurs in response to numerous increased afterload stimuli and precedes irreversible heart failure (HF). Therefore, therapies that ameliorate pathological cardiac hypertrophy are urgently required. Sirtuin 3 (Sirt3) is a main member of histone deacetylase class III and is a crucial anti-oxidative stress agent. Therapeutically enhancing the Sirt3 transfection efficiency in the heart would broaden the potential clinical application of Sirt3. Ultrasound-targeted microbubble destruction (UTMD) is a prospective, noninvasive, repeatable, and targeted gene delivery technique. In the present study, we explored the potential and safety of UTMD as a delivery tool for Sirt3 in hypertrophic heart tissues using adult male Bama miniature pigs. Pigs were subjected to ear vein delivery of human Sirt3 together with UTMD of cationic microbubbles (CMBs). Fluorescence imaging, western blotting, and quantitative real-time PCR revealed that the targeted destruction of ultrasonic CMBs in cardiac tissues greatly boosted Sirt3 delivery. Overexpression of Sirt3 ameliorated oxidative stress and partially improved the diastolic function and prevented the apoptosis and profibrotic response. Lastly, our data revealed that Sirt3 may regulate the potential transcription of catalase and MnSOD through Foxo3a. Combining the advantages of ultrasound CMBs with preclinical hypertrophy large animal models for gene delivery, we established a classical hypertrophy model as well as a strategy for the targeted delivery of genes to hypertrophic heart tissues. Since oxidative stress, fibrosis and apoptosis are indispensable in the evolution of cardiac hypertrophy and heart failure, our findings suggest that Sirt3 is a promising therapeutic option for these diseases. STATEMENT OF SIGNIFICANCE: : Pathological cardiac hypertrophy is a central prepathology of heart failure and is seen to eventually precede it. Feasible targets that may prevent or reverse disease progression are scarce and urgently needed. In this study, we developed surface-filled lipid octafluoropropane gas core cationic microbubbles that could target the release of human Sirt3 reactivating the endogenous Sirt3 in hypertrophic hearts and protect against oxidative stress in a pig model of cardiac hypertrophy induced by aortic banding. Sirt3-CMBs may enhance cardiac diastolic function and ameliorate fibrosis and apoptosis. Our work provides a classical cationic lipid-based, UTMD-mediated Sirt3 delivery system for the treatment of Sirt3 in patients with established cardiac hypertrophy, as well as a promising therapeutic target to combat pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Xionghai Qin
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Peian Cai
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Chang Liu
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Kegong Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xingpei Jiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Wei Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Jiarou Li
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xuan Jiao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Erliang Guo
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yixiu Yu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Lu Sun
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hai Tian
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
4
|
Nijholt KT, Voorrips SN, Sánchez-Aguilera PI, Westenbrink BD. Exercising heart failure patients: cardiac protection through preservation of mitochondrial function and substrate utilization? CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
5
|
Li K, Cardoso C, Moctezuma-Ramirez A, Elgalad A, Perin E. Evaluation of large animal models for preclinical studies of heart failure with preserved ejection fraction using clinical score systems. Front Cardiovasc Med 2023; 10:1099453. [PMID: 37034319 PMCID: PMC10076838 DOI: 10.3389/fcvm.2023.1099453] [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: 11/15/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a complex, heterogeneous spectrum of pathologic features combined with average left ventricular volume and diastolic dysfunction. HFpEF is a significant public health problem associated with high morbidity and mortality rates. Currently, effective treatments for HFpEF represent the greatest unmet need in cardiovascular medicine. A lack of an efficient preclinical model has hampered the development of new devices and medications for HFpEF. Because large animal models have similar physiologic traits as humans and appropriate organ sizes, they are the best option for limiting practical constraints. HFpEF is a highly integrated, multiorgan, systemic disorder requiring a multipronged investigative approach. Here, we review the large animal models of HFpEF reported to date and describe the methods that have been used to create HFpEF, including surgery-induced pressure overloading, medicine-induced pressure overloading, and diet-induced metabolic syndrome. In addition, for the first time to our knowledge, we use two established clinical HFpEF algorithms (HFA-PEFF and H2FPEF scores) to evaluate the currently available large animal models. We also discuss new technologies, such as continuous remote pressure monitors and inflatable aortic cuffs, as well as how the models could be improved. Based on current progress and our own experience, we believe an efficient large animal model of HFpEF should simultaneously encompass multiple pathophysiologic factors, along with multiorgan dysfunction. This could be fully evaluated through available methods (imaging, blood work). Although many models have been studied, only a few studies completely meet clinical score standards. Therefore, it is critical to address the deficiencies of each model and incorporate novel techniques to establish a more reliable model, which will help facilitate the understanding of HFpEF mechanisms and the development of a treatment.
Collapse
Affiliation(s)
- Ke Li
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Cristiano Cardoso
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Angel Moctezuma-Ramirez
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Abdelmotagaly Elgalad
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
- Correspondence: Abdelmotagaly Elgalad
| | - Emerson Perin
- Center for Clinical Research, The Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
6
|
Zaid M, Sala L, Ivey JR, Tharp DL, Mueller CM, Thorne PK, Kelly SC, Silva KAS, Amin AR, Ruiz-Lozano P, Kapiloff MS, Despins L, Popescu M, Keller J, Skubic M, Ahmad S, Emter CA, Guidoboni G. Mechanism-Driven Modeling to Aid Non-invasive Monitoring of Cardiac Function via Ballistocardiography. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:788264. [PMID: 35252962 PMCID: PMC8888976 DOI: 10.3389/fmedt.2022.788264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022] Open
Abstract
Left ventricular (LV) catheterization provides LV pressure-volume (P-V) loops and it represents the gold standard for cardiac function monitoring. This technique, however, is invasive and this limits its applicability in clinical and in-home settings. Ballistocardiography (BCG) is a good candidate for non-invasive cardiac monitoring, as it is based on capturing non-invasively the body motion that results from the blood flowing through the cardiovascular system. This work aims at building a mechanistic connection between changes in the BCG signal, changes in the P-V loops and changes in cardiac function. A mechanism-driven model based on cardiovascular physiology has been used as a virtual laboratory to predict how changes in cardiac function will manifest in the BCG waveform. Specifically, model simulations indicate that a decline in LV contractility results in an increase of the relative timing between the ECG and BCG signal and a decrease in BCG amplitude. The predicted changes have subsequently been observed in measurements on three swine serving as pre-clinical models for pre- and post-myocardial infarction conditions. The reproducibility of BCG measurements has been assessed on repeated, consecutive sessions of data acquisitions on three additional swine. Overall, this study provides experimental evidence supporting the utilization of mechanism-driven mathematical modeling as a guide to interpret changes in the BCG signal on the basis of cardiovascular physiology, thereby advancing the BCG technique as an effective method for non-invasive monitoring of cardiac function.
Collapse
Affiliation(s)
- Mohamed Zaid
- Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, MO, United States
| | - Lorenzo Sala
- Centre de Recherche Inria Saclay Île-De-France, Palaiseau, France
| | - Jan R. Ivey
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Darla L. Tharp
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Christina M. Mueller
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Pamela K. Thorne
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Shannon C. Kelly
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Kleiton Augusto Santos Silva
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Amira R. Amin
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | | | - Michael S. Kapiloff
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, United States
| | - Laurel Despins
- Sinclair School of Nursing, University of Missouri, Columbia, MO, United States
| | - Mihail Popescu
- Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO, United States
| | - James Keller
- Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, MO, United States
| | - Marjorie Skubic
- Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, MO, United States
| | - Salman Ahmad
- Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Giovanna Guidoboni
- Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, MO, United States
- Mathematics, College of Arts and Science, University of Missouri, Columbia, MO, United States
- *Correspondence: Giovanna Guidoboni
| |
Collapse
|
7
|
Miyagi C, Miyamoto T, Kuroda T, Karimov JH, Starling RC, Fukamachi K. Large animal models of heart failure with preserved ejection fraction. Heart Fail Rev 2021; 27:595-608. [PMID: 34751846 DOI: 10.1007/s10741-021-10184-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/14/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by diastolic dysfunction and multiple comorbidities. The number of patients is continuously increasing, with no improvement in its unfavorable prognosis, and there is a strong need for novel treatments. New devices and drugs are difficult to assess at the translational preclinical step due to the lack of high-fidelity large animal models of HFpEF. In this review, we describe the summary of historical and evolving techniques for developing large animal models. The representative methods are pressure overload models, including (1) aortic banding, (2) aortic stent, (3) renal hypertension, and (4) mineralocorticoid-induced hypertension. Diet-induced metabolic syndromes are also used. A new technique with an inflatable balloon inside the left ventricle can be used during acute/chronic in vivo surgeries to simulate HFpEF-like hemodynamics for pump-based therapies. Canines and porcine are most widely used, but other non-rodent animals (sheep, non-human primates, felines, or calves) have been used. Feline models present the most well-simulated HFpEF pathology, but small size is a concern, and the information is still very limited. The rapid and reliable establishment of large animal models for HFpEF, and novel methodology based on the past experimental attempts with large animals, are needed.
Collapse
Affiliation(s)
- Chihiro Miyagi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Takuma Miyamoto
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Taiyo Kuroda
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Jamshid H Karimov
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Randall C Starling
- Department of Cardiovascular Medicine, Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA.,Kaufman Center for Heart Failure Treatment and Recovery, Cleveland Clinic, Cleveland, OH, USA
| | - Kiyotaka Fukamachi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
8
|
Oliveira MI, Leite S, Barros A, Lourenço AP, Mendes C, Schmidt C, Santos M, Leite-Moreira A, Moreira-Gonçalves D. Histological and haemodynamic characterization of right ventricle in sedentary and trained rats with heart failure with preserved ejection fraction. Exp Physiol 2021; 106:2457-2471. [PMID: 34676608 DOI: 10.1113/ep089516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/12/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Right ventricle (RV) dysfunction is highly prevalent in heart failure with preserved ejection fraction (HFpEF), nearly doubling the risk of death: what are the RV functional and structural changes in HFpEF and how does aerobic exercise impact them? What is the main finding and its importance? The HFpEF ZSF1 rat model presents RV structural and functional changes mimicking the human condition. Aerobic exercise prevented the decline in V ̇ O 2 max , lowered surrogate markers of RV overload (e.g., higher mean and maximum systolic pressure) and improved diastolic dysfunction (e.g., end-diastolic pressure and relaxation time constant). This emphasizes the importance of using exercise to manage HFpEF. ABSTRACT Right ventricle (RV) dysfunction is highly prevalent in heart failure with preserved ejection fraction (HFpEF) and is a marker of poor prognosis. We assessed the obese ZSF1 rat model of HFpEF to ascertain if these animals also develop RV dysfunction and evaluated whether aerobic exercise could prevent this. Obese ZSF1 rats were randomly allocated to an aerobic exercise training group (n = 7; treadmill running, 5 days/week, 60 min/day, 15 m/min for 5 weeks) or to a sedentary group (n = 7). We used lean ZSF1 rats (n = 7) as the control group. After 5 weeks, rats were submitted to an exercise tolerance test and invasive haemodynamic evaluation, killed and samples from the RV collected for histological analysis. Obese sedentary ZSF1 rats showed lower V ̇ O 2 max , RV pressure overload (e.g., higher mean and maximum systolic pressure) and diastolic dysfunction (e.g., higher minimum and end-diastolic pressure and relaxation time constant), paralleled by RV cardiomyocyte hypertrophy. Except for cardiomyocyte hypertrophy, aerobic exercise prevented these functional changes. Our data support that this model of HFpEF shows functional and structural changes in the RV that resemble the human HFpEF phenotype, reinforcing its utility to understand this pathophysiology and to adress novel therapeutic targets to manage HFpEF. In addition, we showed that aerobic exercise is cardioprotective for the RV. A deeper knowledge of the mechanisms underlying the benefits of aerobic exercise could also lead to the identification of therapeutic targets to be further explored.
Collapse
Affiliation(s)
- Maria Isilda Oliveira
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Centre of Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, Porto, Portugal
| | - Sara Leite
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,USF Anta, ACeS Gaia/Espinho, Porto, Portugal
| | - António Barros
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - André P Lourenço
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Anesthesiology, São João University Hospital, Porto, Portugal
| | - Cláudia Mendes
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Cristine Schmidt
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Centre of Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, Porto, Portugal
| | - Mário Santos
- CardioVascular Research Group, Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Cardiology Department, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Adelino Leite-Moreira
- Cardiovascular R&D Center (UnIC) and Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Cardiothoracic Surgery, São João University Hospital, Porto, Portugal
| | - Daniel Moreira-Gonçalves
- Centre of Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
9
|
Jaconiano E, Moreira-Gonçalves D. Unveiling the role of exercise training in targeting the inflammatory paradigm of heart failure with preserved ejection fraction: a narrative review. Heart Fail Rev 2021; 27:163-190. [PMID: 34244870 DOI: 10.1007/s10741-021-10138-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 12/30/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is currently lacking an effective pharmacological treatment with impact on major outcomes such as hospitalization and mortality. Exercise training (EXT) is recognized as an important nonpharmacological tool, capable of improving exercise capacity and quality of life, and has even been associated with a reduction in hospitalization and cardiovascular mortality risk. However, this positive impact largely lacks a physiological explanation. The aim of this narrative review was to provide an overview of the available data supporting the hypothesis that the beneficial role of EXT in HFpEF might be due to its effects on targeting the inflammatory paradigm described for this disease. A comprehensive literature search was conducted using the PubMed-NCBI database. We reviewed the effects of EXT throughout each step of the pathophysiological pathway leading to HFpEF and found clinical and/or preclinical evidence supporting the reduction of systemic inflammation, endothelial dysfunction, microvascular rarefaction, and myocardial stiffness. We also highlighted some gaps in the knowledge or topics that deserve further clarification in future studies. In conclusion, despite the scarcity of clinical studies in this population, there is compelling evidence suggesting that EXT modulates crucial aspects of the inflammatory pathway described for HFpEF and future investigation on cellular and molecular mechanisms are encouraged.
Collapse
Affiliation(s)
- Eliane Jaconiano
- Cardiovascular R&D Center (UnIC) and Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Daniel Moreira-Gonçalves
- Centre of Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Kelly SC, Rau CD, Ouyang A, Thorne PK, Olver TD, Edwards JC, Domeier TL, Padilla J, Grisanti LA, Fleenor BS, Wang Y, Rector RS, Emter CA. The right ventricular transcriptome signature in Ossabaw swine with cardiometabolic heart failure: implications for the coronary vasculature. Physiol Genomics 2021; 53:99-115. [PMID: 33491589 PMCID: PMC7988741 DOI: 10.1152/physiolgenomics.00093.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) patients with deteriorating right ventricular (RV) structure and function have a nearly twofold increased risk of death compared with those without. Despite the well-established clinical risk, few studies have examined the molecular signature associated with this HF condition. The purpose of this study was to integrate morphological, molecular, and functional data with the transcriptome data set in the RV of a preclinical model of cardiometabolic HF. Ossabaw swine were fed either normal diet without surgery (lean control, n = 5) or Western diet and aortic-banding (WD-AB; n = 4). Postmortem RV weight was increased and positively correlated with lung weight in the WD-AB group compared with CON. Total RNA-seq was performed and gene expression profiles were compared and analyzed using principal component analysis, weighted gene co-expression network analysis, module enrichment analysis, and ingenuity pathway analysis. Gene networks specifically associated with RV hypertrophic remodeling identified a hub gene in MAPK8 (or JNK1) that was associated with the selective induction of the extracellular matrix (ECM) component fibronectin. JNK1 and fibronectin protein were increased in the right coronary artery (RCA) of WD-AB animals and associated with a decrease in matrix metalloproteinase 14 protein, which specifically degrades fibronectin. RCA fibronectin content was correlated with increased vascular stiffness evident as a decreased elastin elastic modulus in WD-AB animals. In conclusion, this study establishes a molecular and transcriptome signature in the RV using Ossabaw swine with cardiometabolic HF. This signature was associated with altered ECM regulation and increased vascular stiffness in the RCA, with selective dysregulation of fibronectin.
Collapse
Affiliation(s)
- Shannon C Kelly
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Christoph D Rau
- Department of Computational Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - An Ouyang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pamela K Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jenna C Edwards
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Laurel A Grisanti
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Bradley S Fleenor
- Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, Indiana
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, California
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine-Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial VA Hospital, University of Missouri, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
11
|
Magnusson JL, Emter CA, Cummings KJ. Sex- and age-based differences in the effect of central serotonin on arterial blood pressure regulation. J Appl Physiol (1985) 2020; 129:1310-1323. [PMID: 32909922 DOI: 10.1152/japplphysiol.00414.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Medullary serotonin (5-hydroxytryptamine; 5-HT) neurons project to multiple autonomic nuclei in the central nervous system (CNS). Infant rats lacking 5-HT have low arterial blood pressure (ABP) in quiet sleep, but the role of 5-HT in ABP regulation across vigilance states in adults has not been studied. We hypothesized that in adults, CNS 5-HT deficiency leads to hypotension mainly in quiet wakefulness (QW) and non-rapid eye movement (NREM) sleep, when 5-HT neurons are active. We tested male and female tryptophan hydroxylase 2 knockout rats (TPH2-/-), specifically deficient in CNS 5-HT, and wild-type (TPH2+/+) controls at 2-3, 5-8, and 12-13 mo of age. Compared with TPH2+/+, mean arterial pressure of 5-8- and 12-13-mo-old (middle-aged) male TPH2-/- rats was significantly elevated (∼10 mmHg) in QW and rapid eye movement (REM) sleep. Middle-aged male TPH2-/- rats also had more frequent extreme hypertensive events during prolonged episodes of REM sleep. Female TPH2-/- had normal ABP. The low- and very-low-frequency components of systolic ABP variability were significantly higher in middle-aged male, but not female, TPH2-/- rats compared with in TPH2+/+ rats, suggesting elevated sympathetic vascular tone in male TPH2-/- rats. However, the hypertension of male TPH2-/- rats was not ameliorated by ganglionic blockade. Hearts and lungs of middle-aged male TPH2-/- rats were significantly heavier than those of TPH2+/+ rats. We show that a loss of CNS 5-HT leads to high ABP only in middle-aged males during wakefulness and REM sleep, possibly due to increased vascular tone. It should be investigated whether elevated ventricular afterload associated with CNS 5-HT deficiency initiates cardiac remodeling or alters pulmonary hemodynamics.NEW & NOTEWORTHY The role of serotonin in arterial blood pressure (ABP) regulation across states of vigilance is unknown. We hypothesized that adult rats devoid of CNS serotonin (TPH2-/-) have low ABP in wakefulness and NREM sleep, when serotonin neurons are active. However, TPH2-/- rats experience higher ABP than TPH2+/+ rats in wakefulness and REM only, a phenotype present only in older males and not females. CNS serotonin may be critical for preventing high ABP in males with aging.
Collapse
Affiliation(s)
- Jennifer L Magnusson
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
12
|
Ouyang A, Olver TD, Emter CA, Fleenor BS. Chronic exercise training prevents coronary artery stiffening in aortic-banded miniswine: role of perivascular adipose-derived advanced glycation end products. J Appl Physiol (1985) 2019; 127:816-827. [PMID: 31295062 DOI: 10.1152/japplphysiol.00146.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is associated with increased large conduit artery stiffness and afterload resulting in stiffening of the coronary arteries. Perivascular adipose tissue (PVAT) and advanced glycation end products (AGE) both promote arterial stiffness, yet the mechanisms by which coronary PVAT promotes arterial stiffness and the efficacy of exercise to prevent coronary stiffness are unknown. We hypothesized that both chronic continuous and interval exercise training would prevent coronary PVAT-mediated AGE secretion and arterial stiffness. Yucatan miniature swine were divided into four groups: control-sedentary (CON), aortic banded sedentary-heart failure (HF), aortic banded HF-continuous exercise trained (HF+CONT), and aortic banded HF-interval exercise trained (HF+IT). The left circumflex and right coronary arteries underwent ex vivo mechanical testing, and arterial AGE, elastin, and collagen were assessed. Coronary elastin elastic modulus (EEM) and elastin protein were lower and AGE was increased with HF compared with CON, which was prevented by both HF+CONT and HF+IT. Mouse aortic segments treated with swine coronary PVAT conditioned medium had lower EEM and elastin content and greater AGE secretion and arterial AGE accumulation in HF compared with CON, which was prevented by both HF+CONT and HF+IT. Aminoguanidine (AMG), an AGE inhibitor, prevented the reduction in EEM, arterial elastin content, and AGE accumulation in mouse aortic segments treated with PVAT conditioned medium in the HF group. Our data demonstrate efficacy for chronic continuous and interval exercise to prevent coronary artery stiffness via inhibition of PVAT-derived AGE secretion in a preclinical miniswine model of pressure overload-induced HF.NEW & NOTEWORTHY Our findings show that chronic continuous and interval exercise training regimens prevent coronary artery stiffness associated with inhibition of perivascular adipose tissue-derived advanced glycation end products in a translational pressure overload-induced heart failure model potentially providing an effective therapeutic option for heart failure patients.
Collapse
Affiliation(s)
- An Ouyang
- Department of Kinesiology and Health Promotion, University of Kentucky, Lexington, Kentucky
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Bradley S Fleenor
- Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, Indiana
| |
Collapse
|
13
|
Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, Emter CA. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure. JACC Basic Transl Sci 2019; 4:404-421. [PMID: 31312763 PMCID: PMC6610000 DOI: 10.1016/j.jacbts.2019.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
Collapse
Key Words
- AB, aortic-banded
- CON, control
- EDPVR, end-diastolic pressure−volume relationship
- EF, ejection fraction
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL1RL1, interleukin 1 receptor-like 1
- LV, left ventricle
- NF, nuclear factor
- PTX3, pentraxin-3
- WD, Western Diet
- cardio-metabolic disease
- heart failure
- integrative pathophysiology
- preclinical model of cardiovascular disease
Collapse
Affiliation(s)
- T. Dylan Olver
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Jenna C. Edwards
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Thomas J. Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - John L. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Chen Gao
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christoph Rau
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chad M. Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Paula J. Klutho
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Linda Alex
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | | | - Jan R. Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Pamela K. Thorne
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Christopher P. Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A. Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University- School of Medicine, St. Louis, Missouri
| | - Timothy L. Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
- Department of Child Health, University of Missouri-Columbia, Columbia, Missouri
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Department of Medicine – University of Missouri-Columbia, Columbia, Missouri
- Research Service, Harry S Truman Memorial VA Hospital, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A. Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
14
|
Adams V, Linke A. Impact of exercise training on cardiovascular disease and risk. Biochim Biophys Acta Mol Basis Dis 2019; 1865:728-734. [DOI: 10.1016/j.bbadis.2018.08.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/03/2018] [Accepted: 08/15/2018] [Indexed: 01/07/2023]
|
15
|
Kiyuna LA, Albuquerque RPE, Chen CH, Mochly-Rosen D, Ferreira JCB. Targeting mitochondrial dysfunction and oxidative stress in heart failure: Challenges and opportunities. Free Radic Biol Med 2018; 129:155-168. [PMID: 30227272 PMCID: PMC6309415 DOI: 10.1016/j.freeradbiomed.2018.09.019] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/28/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction characterized by impaired bioenergetics, oxidative stress and aldehydic load is a hallmark of heart failure. Recently, different research groups have provided evidence that selective activation of mitochondrial detoxifying systems that counteract excessive accumulation of ROS, RNS and reactive aldehydes is sufficient to stop cardiac degeneration upon chronic stress, such as heart failure. Therefore, pharmacological and non-pharmacological approaches targeting mitochondria detoxification may play a critical role in the prevention or treatment of heart failure. In this review we discuss the most recent findings on the central role of mitochondrial dysfunction, oxidative stress and aldehydic load in heart failure, highlighting the most recent preclinical and clinical studies using mitochondria-targeted molecules and exercise training as effective tools against heart failure.
Collapse
Affiliation(s)
- Ligia Akemi Kiyuna
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, USA
| | | |
Collapse
|
16
|
Baines CP, Gutiérrez-Aguilar M. The still uncertain identity of the channel-forming unit(s) of the mitochondrial permeability transition pore. Cell Calcium 2018; 73:121-130. [PMID: 29793100 PMCID: PMC5993635 DOI: 10.1016/j.ceca.2018.05.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/07/2018] [Accepted: 05/12/2018] [Indexed: 10/16/2022]
Abstract
Mitochondria from different organisms can undergo a sudden process of inner membrane unselective leakiness to molecules known as the mitochondrial permeability transition (MPT). This process has been studied for nearly four decades and several proteins have been claimed to constitute, or at least regulate the usually inactive pore responsible for this transition. However, no protein candidate proposed as the actual pore-forming unit has passed rigorous gain- or loss-of-function genetic tests. Here we review evidence for -and against- putative channel-forming components of the MPT pore. We conclude that the structure of the MPT pore still remains largely undefined and suggest that future studies should follow established technical considerations to unambiguously consolidate the channel forming constituent(s) of the MPT pore.
Collapse
Affiliation(s)
- Christopher P Baines
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211, USA; Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211, USA.
| | - Manuel Gutiérrez-Aguilar
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Ciudad de México, Mexico.
| |
Collapse
|
17
|
Fleenor BS, Ouyang A, Olver TD, Hiemstra JA, Cobb MS, Minervini G, Emter CA. Saxagliptin Prevents Increased Coronary Vascular Stiffness in Aortic-Banded Mini Swine. Hypertension 2018; 72:466-475. [PMID: 29891647 DOI: 10.1161/hypertensionaha.118.10993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/25/2018] [Accepted: 05/14/2018] [Indexed: 01/09/2023]
Abstract
Increased peripheral conduit artery stiffness has been shown in patients with heart failure (HF) with preserved ejection fraction. However, it is unknown whether this phenomenon extends to the coronary vasculature. HF with preserved ejection fraction may be driven, in part, by coronary inflammation, and inhibition of the enzyme DPP-4 (dipeptidyl-peptidase 4) reduces inflammation and oxidative stress. The purpose of this study was to determine the effect of saxagliptin-a DPP-4 inhibitor-on coronary stiffness in aortic-banded mini swine. We hypothesized saxagliptin would prevent increased coronary artery stiffness in a translational swine model with cardiac features of HF with preserved ejection fraction by inhibiting perivascular adipose tissue inflammation. Yucatan mini swine were divided into 3 groups: control, aortic-banded untreated HF, and aortic-banded saxagliptin-treated HF. Ex vivo mechanical testing was performed on the left circumflex and right coronary arteries, and advanced glycation end product, NF-κB (nuclear factor-κB), and nitrotyrosine levels were measured. An increase in the coronary elastic modulus of HF animals was associated with increased vascular advanced glycation end products, NF-κB, and nitrotyrosine levels compared with control and prevented by saxagliptin treatment. Aortas from healthy mice were treated with media from swine perivascular adipose tissue culture to assess its role on vascular stiffening. Conditioned media from HF and saxagliptin-treated HF animals increased mouse aortic stiffness; however, only perivascular adipose tissue from the HF group showed increased advanced glycation end products and NF-κB levels. In conclusion, our data show increased coronary conduit vascular stiffness was prevented by saxagliptin and associated with decreased advanced glycation end products, NF-κB, and nitrotyrosine levels in a swine model with potential relevance to HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Bradley S Fleenor
- From the Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, IN (B.S.F.)
| | - An Ouyang
- Department of Kinesiology and Health Promotion, University of Kentucky, Lexington (A.O.)
| | - T Dylan Olver
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | - Jessica A Hiemstra
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | - Melissa S Cobb
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | | | - Craig A Emter
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| |
Collapse
|
18
|
Olver TD, Edwards JC, Ferguson BS, Hiemstra JA, Thorne PK, Hill MA, Laughlin MH, Emter CA. Chronic interval exercise training prevents BK Ca channel-mediated coronary vascular dysfunction in aortic-banded miniswine. J Appl Physiol (1985) 2018; 125:86-96. [PMID: 29596016 DOI: 10.1152/japplphysiol.01138.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Conventional treatments have failed to improve the prognosis of heart failure with preserved ejection fraction (HFpEF) patients. Thus, the purpose of this study was to determine the therapeutic efficacy of chronic interval exercise training (IT) on large-conductance Ca2+-activated K+ (BKCa) channel-mediated coronary vascular function in heart failure. We hypothesized that chronic interval exercise training would attenuate pressure overload-induced impairments to coronary BKCa channel-mediated function. A translational large-animal model with cardiac features of HFpEF was used to test this hypothesis. Specifically, male Yucatan miniswine were divided into three groups ( n = 7/group): control (CON), aortic banded (AB)-heart failure (HF), and AB-interval trained (HF-IT). Coronary blood flow, vascular conductance, and vasodilatory capacity were measured after administration of the BKCa channel agonist NS-1619 both in vivo and in vitro in the left anterior descending coronary artery and isolated coronary arterioles, respectively. Skeletal muscle citrate synthase activity was decreased and left ventricular brain natriuretic peptide levels increased in HF vs. CON and HF-IT animals. A parallel decrease in NS-1619-dependent coronary vasodilatory reserve in vivo and isolated coronary arteriole vasodilatory responsiveness in vitro were observed in HF animals compared with CON, which was prevented in the HF-IT group. Although exercise training prevented BKCa channel-mediated coronary vascular dysfunction, it did not change BKCa channel α-subunit mRNA, protein, or cellular location (i.e., membrane vs. cytoplasm). In conclusion, these results demonstrate the viability of chronic interval exercise training as a therapy for central and peripheral adaptations of experimental heart failure, including BKCa channel-mediated coronary vascular dysfunction. NEW & NOTEWORTHY Conventional treatments have failed to improve the prognosis of heart failure with preserved ejection fraction (HFpEF) patients. Our findings show that chronic interval exercise training can prevent BKCa channel-mediated coronary vascular dysfunction in a translational swine model of chronic pressure overload-induced heart failure with relevance to human HFpEF.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| | - Jenna C Edwards
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| | - Brian S Ferguson
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| | - Jessica A Hiemstra
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| | - Pamela K Thorne
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri-Columbia , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia , Columbia, Missouri
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri-Columbia , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia , Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia , Columbia, Missouri
| |
Collapse
|
19
|
Hassanpour SH, Dehghani MA, Karami SZ. Study of respiratory chain dysfunction in heart disease. J Cardiovasc Thorac Res 2018; 10:1-13. [PMID: 29707171 PMCID: PMC5913686 DOI: 10.15171/jcvtr.2018.01] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/25/2017] [Indexed: 02/06/2023] Open
Abstract
The relentlessly beating heart has the greatest oxygen consumption of any organ in the body at rest reflecting its huge metabolic turnover and energetic demands. The vast majority of its energy is produced and cycled in form of ATP which stems mainly from oxidative phosphorylation occurring at the respiratory chain in the mitochondria. A part from energy production, the respiratory chain is also the main source of reactive oxygen species and plays a pivotal role in the regulation of oxidative stress. Dysfunction of the respiratory chain is therefore found in most common heart conditions. The pathophysiology of mitochondrial respiratory chain dysfunction in hereditary cardiac mitochondrial disease, the aging heart, in LV hypertrophy and heart failure, and in ischaemia-reperfusion injury is reviewed. We introduce the practicing clinician to the complex physiology of the respiratory chain, highlight its impact on common cardiac disorders and review translational pharmacological and non-pharmacological treatment strategies.
Collapse
Affiliation(s)
| | - Mohammad Amin Dehghani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
20
|
Yin G, Yang X, Li Q, Guo Z. GATA1 activated lncRNA (Galont) promotes anoxia/reoxygenation-induced autophagy and cell death in cardiomyocytes by sponging miR-338. J Cell Biochem 2018; 119:4161-4169. [PMID: 29247537 DOI: 10.1002/jcb.26623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022]
Abstract
The hypernomic autophagy is associated with various cardiovascular diseases. Long noncoding RNAs (lncRNAs) are emerging as important regulators in gene expression, which have been involved in multiple physiological and pathological processes. However, the function of lncRNAs and how they functioned in the autophagy in cardiomyocytes were rarely reported. In this study, we report that a lncRNA, named GATA1 activated lncRNA (Galont), can directly interact with miR-338 and promote ATG5-mediated autophagic cell death in murine cardiomyocytes. First, we found that Galont was upregulated by anoxia/reoxygenation (A/R) stimulus, and it was able to promote autophagy and cell death in cardiomyocytes exposure to A/R. Then, miR-338 was identified as a novel suppressor in autophagy and autophagic cell death. Our results from bioinformatic analysis and luciferase reporter gene assay indicated that ATG5 is a target gene of miR-338. Furthermore, RNA pull-down assays demonstrated that Galont directly interacted with miR-338, and thus promoted ATG5 expression and autophagic cell death. Our findings reveal a novel regulatory circuit in the autophagy in cardiomyocytes, which consists of Galont, miR-338 and ATG5.
Collapse
Affiliation(s)
- Guotian Yin
- Department of Cardiology, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Xiuli Yang
- Department of Cardiology, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
21
|
Hiemstra JA, Veteto AB, Lambert MD, Olver TD, Ferguson BS, McDonald KS, Emter CA, Domeier TL. Chronic low-intensity exercise attenuates cardiomyocyte contractile dysfunction and impaired adrenergic responsiveness in aortic-banded mini-swine. J Appl Physiol (1985) 2018; 124:1034-1044. [PMID: 29357490 DOI: 10.1152/japplphysiol.00840.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Exercise improves clinical outcomes in patients diagnosed with heart failure with reduced ejection fraction (HFrEF), in part via beneficial effects on cardiomyocyte Ca2+ cycling during excitation-contraction coupling (ECC). However, limited data exist regarding the effects of exercise training on cardiomyocyte function in patients diagnosed with heart failure with preserved ejection fraction (HFpEF). The purpose of this study was to investigate cardiomyocyte Ca2+ handling and contractile function following chronic low-intensity exercise training in aortic-banded miniature swine and test the hypothesis that low-intensity exercise improves cardiomyocyte function in a large animal model of pressure overload. Animals were divided into control (CON), aortic-banded sedentary (AB), and aortic-banded low-intensity trained (AB-LIT) groups. Left ventricular cardiomyocytes were electrically stimulated (0.5 Hz) to assess Ca2+ homeostasis (fura-2-AM) and unloaded shortening during ECC under conditions of baseline pacing and pacing with adrenergic stimulation using dobutamine (1 μM). Cardiomyocytes in AB animals exhibited depressed Ca2+ transient amplitude and cardiomyocyte shortening vs. CON under both conditions. Exercise training attenuated AB-induced decreases in cardiomyocyte Ca2+ transient amplitude but did not prevent impaired shortening vs. CON. With dobutamine, AB-LIT exhibited both Ca2+ transient and shortening amplitude similar to CON. Adrenergic sensitivity, assessed as the time to maximum inotropic response following dobutamine treatment, was depressed in the AB group but normal in AB-LIT animals. Taken together, our data suggest exercise training is beneficial for cardiomyocyte function via the effects on Ca2+ homeostasis and adrenergic sensitivity in a large animal model of pressure overload-induced heart failure. NEW & NOTEWORTHY Conventional treatments have failed to improve the prognosis of heart failure with preserved ejection fraction (HFpEF) patients. Our findings show chronic low-intensity exercise training can prevent cardiomyocyte dysfunction and impaired adrenergic responsiveness in a translational large animal model of chronic pressure overload-induced heart failure with relevance to human HFpEF.
Collapse
Affiliation(s)
- Jessica A Hiemstra
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Missouri , Columbia, Missouri
| | - Adam B Veteto
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri , Columbia, Missouri
| | - Michelle D Lambert
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri , Columbia, Missouri
| | - T Dylan Olver
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Missouri , Columbia, Missouri
| | - Brian S Ferguson
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Missouri , Columbia, Missouri
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri , Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Missouri , Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri , Columbia, Missouri
| |
Collapse
|
22
|
Lum-Naihe K, Toedebusch R, Mahmood A, Bajwa J, Carmack T, Kumar SA, Ardhanari S, DeMarco VG, Emter CA, Pulakat L. Cardiovascular disease progression in female Zucker Diabetic Fatty rats occurs via unique mechanisms compared to males. Sci Rep 2017; 7:17823. [PMID: 29259233 PMCID: PMC5736602 DOI: 10.1038/s41598-017-18003-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/30/2017] [Indexed: 02/08/2023] Open
Abstract
Population studies have shown that compared to diabetic men, diabetic women are at a higher risk of cardiovascular disease. However, the mechanisms underlying this gender disparity are unclear. Our studies in young murine models of type 2 diabetes mellitus (T2DM) and cardiovascular disease show that diabetic male rats develop increased cardiac fibrosis and suppression of intracardiac anti-fibrotic cytokines, while premenopausal diabetic female rats do not. This protection from cardiac fibrosis in female rats can be an estrogen-related effect. However, diabetic female rats develop early subclinical myocardial deformation, cardiac hypertrophy via elevated expression of pro-hypertrophic miR-208a, myocardial damage, and suppression of cardio-reparative Angiotensin II receptor 2 (Agtr2). Diabetic rats of both sexes exhibit a reduction in cardiac capillary density. However, diabetic female rats have reduced expression of neuropilin 1 that attenuates cardiomyopathy compared to diabetic male rats. A combination of cardiac hypertrophy and reduced capillary density likely contributed to increased myocardial structural damage in diabetic female rats. We propose expansion of existing cardiac assessments in diabetic female patients to detect myocardial deformation, cardiac hypertrophy and capillary density via non-invasive imaging, as well as suggest miR-208a, AT2R and neuropilin 1 as potential therapeutic targets and mechanistic biomarkers for cardiac disease in females.
Collapse
Affiliation(s)
- Kelly Lum-Naihe
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Ryan Toedebusch
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65201, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Abuzar Mahmood
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65201, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Jamal Bajwa
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Terry Carmack
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Senthil A Kumar
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA
| | - Sivakumar Ardhanari
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA
| | - Vincent G DeMarco
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, 1600 E Rollins, Columbia, MO, 65201, USA.,Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65201, USA
| | - Lakshmi Pulakat
- Department of Medicine, University of Missouri, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Nutrition and Exercise Physiology, Universtiy of Missouri, 204 Gwynn Hall, Columbia, MO, 65211, USA. .,Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65201, USA. .,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA.
| |
Collapse
|
23
|
Campos JC, Queliconi BB, Bozi LHM, Bechara LRG, Dourado PMM, Andres AM, Jannig PR, Gomes KMS, Zambelli VO, Rocha-Resende C, Guatimosim S, Brum PC, Mochly-Rosen D, Gottlieb RA, Kowaltowski AJ, Ferreira JCB. Exercise reestablishes autophagic flux and mitochondrial quality control in heart failure. Autophagy 2017; 13:1304-1317. [PMID: 28598232 PMCID: PMC5584854 DOI: 10.1080/15548627.2017.1325062] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
We previously reported that facilitating the clearance of damaged mitochondria through macroautophagy/autophagy protects against acute myocardial infarction. Here we characterize the impact of exercise, a safe strategy against cardiovascular disease, on cardiac autophagy and its contribution to mitochondrial quality control, bioenergetics and oxidative damage in a post-myocardial infarction-induced heart failure animal model. We found that failing hearts displayed reduced autophagic flux depicted by accumulation of autophagy-related markers and loss of responsiveness to chloroquine treatment at 4 and 12 wk after myocardial infarction. These changes were accompanied by accumulation of fragmented mitochondria with reduced O2 consumption, elevated H2O2 release and increased Ca2+-induced mitochondrial permeability transition pore opening. Of interest, disruption of autophagic flux was sufficient to decrease cardiac mitochondrial function in sham-treated animals and increase cardiomyocyte toxicity upon mitochondrial stress. Importantly, 8 wk of exercise training, starting 4 wk after myocardial infarction at a time when autophagy and mitochondrial oxidative capacity were already impaired, improved cardiac autophagic flux. These changes were followed by reduced mitochondrial number:size ratio, increased mitochondrial bioenergetics and better cardiac function. Moreover, exercise training increased cardiac mitochondrial number, size and oxidative capacity without affecting autophagic flux in sham-treated animals. Further supporting an autophagy mechanism for exercise-induced improvements of mitochondrial bioenergetics in heart failure, acute in vivo inhibition of autophagic flux was sufficient to mitigate the increased mitochondrial oxidative capacity triggered by exercise in failing hearts. Collectively, our findings uncover the potential contribution of exercise in restoring cardiac autophagy flux in heart failure, which is associated with better mitochondrial quality control, bioenergetics and cardiac function.
Collapse
Affiliation(s)
- Juliane C Campos
- a Department of Anatomy , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
- b The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Bruno B Queliconi
- c Departamento de Bioquímica , Instituto de Química, Universidade de São Paulo , Sao Paulo , Brazil
| | - Luiz H M Bozi
- a Department of Anatomy , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Luiz R G Bechara
- a Department of Anatomy , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | | | - Allen M Andres
- b The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Paulo R Jannig
- e School of Physical Education and Sport , University of Sao Paulo , Sao Paulo , Brazil
| | - Kátia M S Gomes
- a Department of Anatomy , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | | | - Cibele Rocha-Resende
- g Department of Physiology and Biophysics , Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Silvia Guatimosim
- g Department of Physiology and Biophysics , Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Patricia C Brum
- e School of Physical Education and Sport , University of Sao Paulo , Sao Paulo , Brazil
| | - Daria Mochly-Rosen
- h Department of Chemical and Systems Biology , Stanford University School of Medicine , Stanford , CA , USA
| | - Roberta A Gottlieb
- b The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Alicia J Kowaltowski
- c Departamento de Bioquímica , Instituto de Química, Universidade de São Paulo , Sao Paulo , Brazil
| | - Julio C B Ferreira
- a Department of Anatomy , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| |
Collapse
|
24
|
Lalande S, Mueller PJ, Chung CS. The link between exercise and titin passive stiffness. Exp Physiol 2017; 102:1055-1066. [PMID: 28762234 DOI: 10.1113/ep086275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/11/2017] [Indexed: 12/27/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review focuses on how in vivo and molecular measurements of cardiac passive stiffness can predict exercise tolerance and how exercise training can reduce cardiac passive stiffness. What advances does it highlight? This review highlights advances in understanding the relationship between molecular (titin-based) and in vivo (left ventricular) passive stiffness, how passive stiffness modifies exercise tolerance, and how exercise training may be therapeutic for cardiac diseases with increased passive stiffness. Exercise can help alleviate the negative effects of cardiovascular disease and cardiovascular co-morbidities associated with sedentary behaviour; this may be especially true in diseases that are associated with increased left ventricular passive stiffness. In this review, we discuss the inverse relationship between exercise tolerance and cardiac passive stiffness. Passive stiffness is the physical property of cardiac muscle to produce a resistive force when stretched, which, in vivo, is measured using the left ventricular end diastolic pressure-volume relationship or is estimated using echocardiography. The giant elastic protein titin is the major contributor to passive stiffness at physiological muscle (sarcomere) lengths. Passive stiffness can be modified by altering titin isoform size or by post-translational modifications. In both human and animal models, increased left ventricular passive stiffness is associated with reduced exercise tolerance due to impaired diastolic filling, suggesting that increased passive stiffness predicts reduced exercise tolerance. At the same time, exercise training itself may induce both short- and long-term changes in titin-based passive stiffness, suggesting that exercise may be a treatment for diseases associated with increased passive stiffness. Direct modification of passive stiffness to improve exercise tolerance is a potential therapeutic approach. Titin passive stiffness itself may be a treatment target based on the recent discovery of RNA binding motif 20, which modifies titin isoform size and passive stiffness. Translating these discoveries that link exercise and left ventricular passive stiffness may provide new methods to enhance exercise tolerance and treat patients with cardiovascular disease.
Collapse
Affiliation(s)
- Sophie Lalande
- Department of Kinesiology & Health Education, The University of Texas at Austin, Austin, TX, USA
| | | | - Charles S Chung
- Department of Physiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
25
|
Hanft LM, Emter CA, McDonald KS. Cardiac myofibrillar contractile properties during the progression from hypertension to decompensated heart failure. Am J Physiol Heart Circ Physiol 2017; 313:H103-H113. [PMID: 28455288 PMCID: PMC5538866 DOI: 10.1152/ajpheart.00069.2017] [Citation(s) in RCA: 14] [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: 02/02/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 11/22/2022]
Abstract
Heart failure arises, in part, from a constellation of changes in cardiac myocytes including remodeling, energetics, Ca2+ handling, and myofibrillar function. However, little is known about the changes in myofibrillar contractile properties during the progression from hypertension to decompensated heart failure. The aim of the present study was to provide a comprehensive assessment of myofibrillar functional properties from health to heart disease. A rodent model of uncontrolled hypertension was used to test the hypothesis that myocytes in compensated hearts exhibit increased force, higher rates of force development, faster loaded shortening, and greater power output; however, with progression to overt heart failure, we predicted marked depression in these contractile properties. We assessed contractile properties in skinned cardiac myocyte preparations from left ventricles of Wistar-Kyoto control rats and spontaneous hypertensive heart failure (SHHF) rats at ~3, ~12, and >20 mo of age to evaluate the time course of myofilament properties associated with normal aging processes compared with myofilaments from rats with a predisposition to heart failure. In control rats, the myofilament contractile properties were virtually unchanged throughout the aging process. Conversely, in SHHF rats, the rate of force development, loaded shortening velocity, and power all increased at ~12 mo and then significantly fell at the >20-mo time point, which coincided with a decrease in left ventricular fractional shortening. Furthermore, these changes occurred independent of changes in β-myosin heavy chain but were associated with depressed phosphorylation of myofibrillar proteins, and the fall in loaded shortening and peak power output corresponded with the onset of clinical signs of heart failure.NEW & NOTEWORTHY This novel study systematically examined the power-generating capacity of cardiac myofilaments during the progression from hypertension to heart disease. Previously undiscovered changes in myofibrillar power output were found and were associated with alterations in myofilament proteins, providing potential new targets to exploit for improved ventricular pump function in heart failure.
Collapse
Affiliation(s)
- Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Craig A Emter
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
26
|
Adams V, Reich B, Uhlemann M, Niebauer J. Molecular effects of exercise training in patients with cardiovascular disease: focus on skeletal muscle, endothelium, and myocardium. Am J Physiol Heart Circ Physiol 2017; 313:H72-H88. [PMID: 28476924 DOI: 10.1152/ajpheart.00470.2016] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
For decades, we have known that exercise training exerts beneficial effects on the human body, and clear evidence is available that a higher fitness level is associated with a lower incidence of suffering premature cardiovascular death. Despite this knowledge, it took some time to also incorporate physical exercise training into the treatment plan for patients with cardiovascular disease (CVD). In recent years, in addition to continuous exercise training, further training modalities such as high-intensity interval training and pyramid training have been introduced for coronary artery disease patients. The beneficial effect for patients with CVD is clearly documented, and during the last years, we have also started to understand the molecular mechanisms occurring in the skeletal muscle (limb muscle and diaphragm) and endothelium, two systems contributing to exercise intolerance in these patients. In the present review, we describe the effects of the different training modalities in CVD and summarize the molecular effects mainly in the skeletal muscle and cardiovascular system.
Collapse
Affiliation(s)
- Volker Adams
- Clinic of Internal Medicine/Cardiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany; and
| | - Bernhard Reich
- University Institute of Sports Medicine, Prevention and Rehabilitation and Research Institute of Molecular Sports Medicine and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Madlen Uhlemann
- Clinic of Internal Medicine/Cardiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany; and
| | - Josef Niebauer
- University Institute of Sports Medicine, Prevention and Rehabilitation and Research Institute of Molecular Sports Medicine and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
27
|
Rocha LADO, Oliveira KS, Migliolo L, Franco OL. Effect of Moderate Exercise on Mitochondrial Proteome in Heart Tissue of Spontaneous Hypertensive Rats. Am J Hypertens 2016; 29:696-704. [PMID: 26391256 DOI: 10.1093/ajh/hpv160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 08/19/2015] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Hypertension is a multifactorial disease and an important independent risk factor for cardiovascular diseases. Exercise training is one of the most important non-pharmacological therapeutic strategies for treating hypertension; however, mitochondrial adaptations in the hypertensive heart as a result of exercise remain obscure. METHODS Aiming to explore the effects of exercise training of moderate intensity on the mitochondrial proteome in hypertensive animal models before and after the pathology developed, 20 isogenic male spontaneous hypertensive rats (SHRs) were randomly divided into 2 groups, 1 with animals of 6 and 40 weeks of age. Animals were submitted to exercise training on a treadmill for 30 minutes, 5 days per week for 4 weeks at 90% of the anaerobic threshold (AT). A mitochondrial sample extract from the left ventricle was prepared and further analyzed using LC-MS/MS. RESULTS Proteomics analyses led to the identification of 143 proteins in all groups. The data showed a considerable and clear increase in the abundance of NADH dehydrogenase and ATP synthase, as well as voltage-dependent anion channel (VDAC) type 1 decrease in exercise groups. When exercise effects were compared, differential proteins expressed only in exercise increased, such as cytochrome c oxidase, alcohol dehydrogenase, and NADH dehydrogenase [ubiquinone] 1 alpha subcomplex. CONCLUSIONS The results support the proposition that moderate exercise induces a beneficial adaptation in left ventricle myocardial mitochondria in order to attenuate the decrease in ATP production in hypertensive models.
Collapse
Affiliation(s)
- Luiz Antonio de Oliveira Rocha
- Curso de Educação Física, Universidade Católica de Brasilia, Brasília, DF, Brazil; Curso de pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil; Centro de Analises Proteomicas e Bioquimicas, Curso de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Kleber Souza Oliveira
- Centro de Analises Proteomicas e Bioquimicas, Curso de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Catolica Dom Bosco, Campo Grande, MS, Brazil
| | - Ludovico Migliolo
- Centro de Analises Proteomicas e Bioquimicas, Curso de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Catolica Dom Bosco, Campo Grande, MS, Brazil
| | - Octavio Luiz Franco
- Curso de pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil; Centro de Analises Proteomicas e Bioquimicas, Curso de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pós-Graduação em Biotecnologia, Universidade Catolica Dom Bosco, Campo Grande, MS, Brazil.
| |
Collapse
|
28
|
Linden MA, Sheldon RD, Meers GM, Ortinau LC, Morris EM, Booth FW, Kanaley JA, Vieira-Potter VJ, Sowers JR, Ibdah JA, Thyfault JP, Laughlin MH, Rector RS. Aerobic exercise training in the treatment of non-alcoholic fatty liver disease related fibrosis. J Physiol 2016; 594:5271-84. [PMID: 27104887 DOI: 10.1113/jp272235] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/13/2016] [Indexed: 12/23/2022] Open
Abstract
KEY POINTS Physiologically relevant rodent models of non-alcoholic steatohepatitis (NASH) that resemble the human condition are limited. Exercise training and energy restriction are first-line recommendations for the treatment of NASH. Hyperphagic Otsuka Long-Evans Tokushima fatty rats fed a western diet high in fat, sucrose and cholesterol for 24 weeks developed a severe NASH with fibrosis phenotype. Moderate intensity exercise training and modest energy restriction provided some improvement in the histological features of NASH that coincided with alterations in markers of hepatic stellate cell activation and extracellular matrix remodelling. The present study highlights the importance of lifestyle modification, including exercise training and energy restriction, in the regulation of advanced liver disease. ABSTRACT The incidence of non-alcoholic steatohepatitis (NASH) is rising but the efficacy of lifestyle modifications to improve NASH-related outcomes remain unclear. We hypothesized that a western diet (WD) would induce NASH in the Otsuka Long-Evans Tokushima Fatty (OLETF) rat and that lifestyle modification would improve this condition. Eight-week-old Long-Evans Tokushima Otsuka (L) and OLETF (O) rats consumed a control diet (10% kcal fat, 3.5% sucrose) or a WD (45% kcal fat, 17% sucrose, 1% cholesterol) for 24 weeks. At 20 weeks of age, additional WD-fed OLETFs were randomized to sedentary (O-SED), food restriction (O-FR; ∼25% kcal reduction vs. O-SED) or exercise training (O-EX; treadmill running 20 m min(-1) with a 15% incline, 60 min day(-1) , 5 days week(-1) ) conditions for 12 weeks. WD induced a NASH phenotype in OLETFs characterized by hepatic fibrosis (collagen 1α1 mRNA and hydroxyproline content), as well as elevated inflammation and non-alcoholic fatty liver disease activity scores, and hepatic stellate cell activation (α-smooth muscle actin) compared to Long-Evans Tokushima Otsuka rats. FR and EX modestly improved NASH-related fibrosis markers (FR: hydroxyproline content, P < 0.01; EX: collagen 1α1 mRNA, P < 0.05; both: fibrosis score, P < 0.01) and inflammation (both: inflammation score; FR: interleukin-1β and tumor necrosis factor α) vs. O-SED. FR reduced hepatic stellate cell activation markers (transforming growth factor-β protein and α-smooth muscle actin mRNA), whereas EX increased the hepatic stellate cell senescence marker CCN1 (P < 0.01 vs. O-SED). Additionally, both FR and EX normalized extracellular matrix remodelling markers to levels similar to L-WD (P > 0.05). Although neither EX nor FR led to complete resolution of the WD-induced NASH phenotype, both independently benefitted liver fibrosis via altered hepatic stellate cell activation and extracellular matrix remodelling.
Collapse
Affiliation(s)
- Melissa A Linden
- Research Service, Harry S Truman Memorial VA Hospital.,Department of Nutrition and Exercise Physiology
| | - Ryan D Sheldon
- Research Service, Harry S Truman Memorial VA Hospital.,Department of Nutrition and Exercise Physiology
| | - Grace M Meers
- Research Service, Harry S Truman Memorial VA Hospital.,Department of Medicine-Division of Gastroenterology and Hepatology
| | | | - E Matthew Morris
- Department of Molecular and Integrative Physiology, University of Kansas Medical Centre, Kansas City, KS, USA
| | - Frank W Booth
- Department of Biomedical Sciences.,Department of Medical Pharmacology and Physiology.,Dalton Cardiovascular Research Centre
| | | | | | - James R Sowers
- Research Service, Harry S Truman Memorial VA Hospital.,Medicine-Division of Endocrinology, University of Missouri, Columbia, MO, USA
| | - Jamal A Ibdah
- Research Service, Harry S Truman Memorial VA Hospital.,Department of Medicine-Division of Gastroenterology and Hepatology.,Department of Medical Pharmacology and Physiology
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Centre, Kansas City, KS, USA.,Kansas City VA Medical Centre, Kansas City, MO, USA
| | | | - R Scott Rector
- Research Service, Harry S Truman Memorial VA Hospital. .,Department of Medicine-Division of Gastroenterology and Hepatology. .,Department of Nutrition and Exercise Physiology.
| |
Collapse
|
29
|
Olver TD, Klakotskaia D, Ferguson BS, Hiemstra JA, Schachtman TR, Laughlin MH, Emter CA. Carotid Artery Vascular Mechanics Serve as Biomarkers of Cognitive Dysfunction in Aortic-Banded Miniature Swine That Can Be Treated With an Exercise Intervention. J Am Heart Assoc 2016; 5:JAHA.116.003248. [PMID: 27207966 PMCID: PMC4889197 DOI: 10.1161/jaha.116.003248] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Cognitive impairment in the setting of heart failure with preserved ejection fraction remains poorly understood. Using aortic‐banded miniature swine displaying pathological features of human heart failure with preserved ejection fraction, we tested the hypothesis that increased carotid artery stiffness and altered carotid blood flow control are associated with impaired memory independent of decreased cardiac output. Furthermore, we hypothesized that chronic exercise prevents carotid artery vascular restructuring and preserves normal blood flow control and cognition in heart failure with preserved ejection fraction. Methods and Results Yucatan pigs aged 8 months were divided into 3 groups: control (n=7), aortic‐banded sedentary (n=7), and aortic‐banded exercise trained (n=7). At 6 months following aortic‐banded or control conditions, memory was evaluated using a spatial hole‐board task. Carotid artery vascular mechanics and blood flow were assessed at rest, and blood flow control was examined during transient vena cava occlusion. Independent of decreased cardiac output, the aortic‐banded group exhibited impaired memory that was associated with carotid artery vascular stiffening, elevated carotid artery vascular resistance, and exaggerated reductions in carotid artery blood flow during vena cava occlusion. Chronic exercise augmented memory scores, normalized blood flow control, and improved indices of carotid artery vascular stiffening. Indices of vascular stiffening were significantly correlated with average memory score. Conclusions Carotid artery stiffness and altered vasomotor control correlate with impaired cognition independent of cardiac systolic dysfunction. Carotid artery vascular mechanics may serve as a biomarker for vascular cognitive impairment in heart failure with preserved ejection fraction. Chronic low‐intensity exercise reduces vascular stiffening and improves cognition, highlighting the utility of exercise therapy for treating vascular cognitive impairment in heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri, Columbia, MO
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - Brian S Ferguson
- Department of Biomedical Sciences, University of Missouri, Columbia, MO
| | | | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri, Columbia, MO Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, MO
| |
Collapse
|
30
|
Hiemstra JA, Lee DI, Chakir K, Gutiérrez-Aguilar M, Marshall KD, Zgoda PJ, Cruz Rivera N, Dozier DG, Ferguson BS, Heublein DM, Burnett JC, Scherf C, Ivey JR, Minervini G, McDonald KS, Baines CP, Krenz M, Domeier TL, Emter CA. Saxagliptin and Tadalafil Differentially Alter Cyclic Guanosine Monophosphate (cGMP) Signaling and Left Ventricular Function in Aortic-Banded Mini-Swine. J Am Heart Assoc 2016; 5:e003277. [PMID: 27098966 PMCID: PMC4843537 DOI: 10.1161/jaha.116.003277] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/03/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cyclic guanosine monophosphate-protein kinase G-phosphodiesterase 5 signaling may be disturbed in heart failure (HF) with preserved ejection fraction, contributing to cardiac remodeling and dysfunction. The purpose of this study was to manipulate cyclic guanosine monophosphate signaling using the dipeptidyl-peptidase 4 inhibitor saxagliptin and phosphodiesterase 5 inhibitor tadalafil. We hypothesized that preservation of cyclic guanosine monophosphate cGMP signaling would attenuate pathological cardiac remodeling and improve left ventricular (LV) function. METHODS AND RESULTS We assessed LV hypertrophy and function at the organ and cellular level in aortic-banded pigs. Concentric hypertrophy was equal in all groups, but LV collagen deposition was increased in only HF animals. Prevention of fibrotic remodeling by saxagliptin and tadalafil was correlated with neuropeptide Y plasma levels. Saxagliptin better preserved integrated LV systolic and diastolic function by maintaining normal LV chamber volumes and contractility (end-systolic pressure-volume relationship, preload recruitable SW) while preventing changes to early/late diastolic longitudinal strain rate. Function was similar to the HF group in tadalafil-treated animals including increased LV contractility, reduced chamber volume, and decreased longitudinal, circumferential, and radial mechanics. Saxagliptin and tadalafil prevented a negative cardiomyocyte shortening-frequency relationship observed in HF animals. Saxagliptin increased phosphodiesterase 5 activity while tadalafil increased cyclic guanosine monophosphate levels; however, neither drug increased downstream PKG activity. Early mitochondrial dysfunction, evident as decreased calcium-retention capacity and Complex II-dependent respiratory control, was present in both HF and tadalafil-treated animals. CONCLUSIONS Both saxagliptin and tadalafil prevented increased LV collagen deposition in a manner related to the attenuation of increased plasma neuropeptide Y levels. Saxagliptin appears superior for treating heart failure with preserved ejection fraction, considering its comprehensive effects on integrated LV systolic and diastolic function.
Collapse
Affiliation(s)
- Jessica A Hiemstra
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | - Dong I Lee
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Khalid Chakir
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Manuel Gutiérrez-Aguilar
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO
| | - Kurt D Marshall
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO
| | - Pamela J Zgoda
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | - Noelany Cruz Rivera
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | - Daniel G Dozier
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | - Brian S Ferguson
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | | | | | - Carolin Scherf
- Department of Veterinary Pathobiology, University of Missouri-Columbia, Columbia, MO
| | - Jan R Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| | | | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO
| | - Christopher P Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO
| | - Craig A Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, MO
| |
Collapse
|
31
|
Rodrigues PG, Leite-Moreira AF, Falcão-Pires I. Myocardial reverse remodeling: how far can we rewind? Am J Physiol Heart Circ Physiol 2016; 310:H1402-22. [PMID: 26993225 DOI: 10.1152/ajpheart.00696.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) is a systemic disease that can be divided into HF with reduced ejection fraction (HFrEF) and with preserved ejection fraction (HFpEF). HFpEF accounts for over 50% of all HF patients and is typically associated with high prevalence of several comorbidities, including hypertension, diabetes mellitus, pulmonary hypertension, obesity, and atrial fibrillation. Myocardial remodeling occurs both in HFrEF and HFpEF and it involves changes in cardiac structure, myocardial composition, and myocyte deformation and multiple biochemical and molecular alterations that impact heart function and its reserve capacity. Understanding the features of myocardial remodeling has become a major objective for limiting or reversing its progression, the latter known as reverse remodeling (RR). Research on HFrEF RR process is broader and has delivered effective therapeutic strategies, which have been employed for some decades. However, the RR process in HFpEF is less clear partly due to the lack of information on HFpEF pathophysiology and to the long list of failed standard HF therapeutics strategies in these patient's outcomes. Nevertheless, new proteins, protein-protein interactions, and signaling pathways are being explored as potential new targets for HFpEF remodeling and RR. Here, we review recent translational and clinical research in HFpEF myocardial remodeling to provide an overview on the most important features of RR, comparing HFpEF with HFrEF conditions.
Collapse
Affiliation(s)
- Patrícia G Rodrigues
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Adelino F Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| |
Collapse
|
32
|
Turner MJ, Guderian S, Wikstrom EA, Huot JR, Peck BD, Arthur ST, Marino JS, Hubbard-Turner T. Altered left ventricular performance in aging physically active mice with an ankle sprain injury. AGE (DORDRECHT, NETHERLANDS) 2016; 38:15. [PMID: 26803818 PMCID: PMC5005884 DOI: 10.1007/s11357-016-9877-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/13/2016] [Indexed: 06/05/2023]
Abstract
We assessed the impact of differing physical activity levels throughout the lifespan, using a musculoskeletal injury model, on the age-related changes in left ventricular (LV) parameters in active mice. Forty male mice (CBA/J) were randomly placed into one of three running wheel groups (transected CFL group, transected ATFL/CFL group, SHAM group) or a SHAM Sedentary group (SHAMSED). Before surgery and every 6 weeks after surgery, LV parameters were measured under 2.5 % isoflurane inhalation. Group effects for daily distance run was significantly greater for the SHAM and lesser for the ATLF/CFL mice (p = 0.013) with distance run decreasing with age for all mice (p < 0.0001). Beginning at 6 months of age, interaction (group × age) was noted with LV posterior wall thickness-to-radius ratios (h/r) where h/r increased with age in the ATFL/CFL and SHAMSED mice while the SHAM and CFL mice exhibited decreased h/r with age (p = 0.0002). Passive filling velocity (E wave) was significantly greater in the SHAM mice and lowest for the ATFL/CFL and SHAMSED mice (p < 0.0001) beginning at 9 months of age. Active filling velocity (A wave) was not different between groups (p = 0.10). Passive-to-active filling velocity ratio (E/A ratio) was different between groups (p < 0.0001), with higher ratios for the SHAM mice and lower ratios for the ATFL/CFL and SHAMSED mice in response to physical activity beginning at 9 months of age. Passive-to-active filling velocity ratio decreased with age (p < 0.0001). Regular physical activity throughout the lifespan improved LV structure, passive filling velocity, and E/A ratio by 6 to 9 months of age and attenuated any negative alterations throughout the second half of life. The diastolic filling differences were found to be significantly related to the amount of activity performed by 9 months and at the end of the lifespan.
Collapse
Affiliation(s)
- Michael J Turner
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Sophie Guderian
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Erik A Wikstrom
- Biodynamics Research Laboratory, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
- Center for Biomedical Engineering & Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Joshua R Huot
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Bailey D Peck
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Susan T Arthur
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Joseph S Marino
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Tricia Hubbard-Turner
- Biodynamics Research Laboratory, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, NC, USA
- Center for Biomedical Engineering & Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
33
|
|
34
|
Alves JP, Nunes RB, Stefani GP, Dal Lago P. Resistance training improves hemodynamic function, collagen deposition and inflammatory profiles: experimental model of heart failure. PLoS One 2014; 9:e110317. [PMID: 25340545 PMCID: PMC4207701 DOI: 10.1371/journal.pone.0110317] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 09/21/2014] [Indexed: 11/30/2022] Open
Abstract
The role of resistance training on collagen deposition, the inflammatory profile and muscle weakness in heart failure remains unclear. Therefore, this study evaluated the influence of a resistance training program on hemodynamic function, maximum strength gain, collagen deposition and inflammatory profile in chronic heart failure rats. Thirty-two male Wistar rats submitted to myocardial infarction by coronary artery ligation or sham surgery were assigned into four groups: sedentary sham (S-Sham, n = 8); trained sham (T-Sham, n = 8); sedentary chronic heart failure (S-CHF, n = 8) and trained chronic heart failure (T-CHF, n = 8). The maximum strength capacity was evaluated by the one maximum repetition test. Trained groups were submitted to an 8-week resistance training program (4 days/week, 4 sets of 10-12 repetitions/session, at 65% to 75% of one maximum repetition). After 8 weeks of the resistance training program, the T-CHF group showed lower left ventricular end diastolic pressure (P<0.001), higher left ventricular systolic pressure (P<0.05), higher systolic blood pressure (P<0.05), an improvement in the maximal positive derivative of ventricular pressure (P<0.05) and maximal negative derivative of ventricular pressure (P<0.05) when compared to the S-CHF group; no differences were observed when compared to Sham groups. In addition, resistance training was able to reduce myocardial hypertrophy (P<0.05), left ventricular total collagen volume fraction (P<0.01), IL-6 (P<0.05), and TNF-α/IL-10 ratio (P<0.05), as well as increasing IL-10 (P<0.05) in chronic heart failure rats when compared to the S-CHF group. Eight weeks of resistance training promotes an improvement of cardiac function, strength gain, collagen deposition and inflammatory profile in chronic heart failure rats.
Collapse
Affiliation(s)
- Jadson P. Alves
- Laboratory of Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Ramiro B. Nunes
- Laboratory of Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Giuseppe P. Stefani
- Laboratory of Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Pedro Dal Lago
- Laboratory of Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Department of Physical Therapy, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
35
|
Hiemstra JA, Gutiérrez-Aguilar M, Marshall KD, McCommis KS, Zgoda PJ, Cruz-Rivera N, Jenkins NT, Krenz M, Domeier TL, Baines CP, Emter CA. A new twist on an old idea part 2: cyclosporine preserves normal mitochondrial but not cardiomyocyte function in mini-swine with compensated heart failure. Physiol Rep 2014; 2:2/6/e12050. [PMID: 24963034 PMCID: PMC4208639 DOI: 10.14814/phy2.12050] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We recently developed a clinically relevant mini‐swine model of heart failure with preserved ejection fraction (HFpEF), in which diastolic dysfunction was associated with increased mitochondrial permeability transition (MPT). Early diastolic function is ATP and Ca2+‐dependent, thus, we hypothesized chronic low doses of cyclosporine (CsA) would preserve mitochondrial function via inhibition of MPT and subsequently maintain normal cardiomyocyte Ca2+ handling and contractile characteristics. Left ventricular cardiomyocytes were isolated from aortic‐banded Yucatan mini‐swine divided into three groups; control nonbanded (CON), HFpEF nontreated (HF), and HFpEF treated with CsA (HF‐CsA). CsA mitigated the deterioration of mitochondrial function observed in HF animals, including functional uncoupling of Complex I‐dependent mitochondrial respiration and increased susceptibility to MPT. Attenuation of mitochondrial dysfunction in the HF‐CsA group was not associated with commensurate improvement in cardiomyocyte Ca2+ handling or contractility. Ca2+ transient amplitude was reduced and transient time to peak and recovery (tau) prolonged in HF and HF‐CsA groups compared to CON. Alterations in Ca2+ transient parameters observed in the HF and HF‐CsA groups were associated with decreased cardiomyocyte shortening and shortening rate. Cellular function was consistent with impaired in vivo systolic and diastolic whole heart function. A significant systemic hypertensive response to CsA was observed in HF‐CsA animals, and may have played a role in the accelerated the development of heart failure at both the whole heart and cellular levels. Given the significant detriment to cardiac function observed in response to CsA, our findings suggest chronic CsA treatment is not a viable therapeutic option for HFpEF. In a recently developed a translational mini‐swine model of heart failure with preserved ejection fraction (HFpEF), we hypothesized inhibiting mitochondrial permeability transition using cyclosporine (CsA) would improve cardiomyocyte function and calcium handling by supporting mitochondrial function. The purpose of this study was to examine the impact of inhibiting cyclophilin D on mitochondrial function and subsequent cardiomyocyte calcium handling using a reduced, nonimmunosuppressive dose of CsA chronically. We found improved mitochondrial function following chronic CsA treatment was not associated with a parallel improvement in cardiomyocyte calcium handling and contractile function, and demonstrate for the first time impaired cardiomyocyte calcium handling and contractile function are present early in the disease process in our HFpEF model.
Collapse
Affiliation(s)
- Jessica A Hiemstra
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Manuel Gutiérrez-Aguilar
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri Dalton Cardiovascular Research Center, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Kurt D Marshall
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri Dalton Cardiovascular Research Center, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Kyle S McCommis
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri Dalton Cardiovascular Research Center, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Pamela J Zgoda
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Noelany Cruz-Rivera
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Nathan T Jenkins
- Department of Kinesiology, University of Georgia, Athens, Georgia
| | - Maike Krenz
- Dalton Cardiovascular Research Center, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri Department of Medical Pharmacology and Physiology, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Christopher P Baines
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri Dalton Cardiovascular Research Center, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Science, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, Missouri
| |
Collapse
|
36
|
Schwarz K, Siddiqi N, Singh S, Neil CJ, Dawson DK, Frenneaux MP. The breathing heart - mitochondrial respiratory chain dysfunction in cardiac disease. Int J Cardiol 2013; 171:134-43. [PMID: 24377708 DOI: 10.1016/j.ijcard.2013.12.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 11/04/2013] [Accepted: 12/11/2013] [Indexed: 01/20/2023]
Abstract
The relentlessly beating heart has the greatest oxygen consumption of any organ in the body at rest reflecting its huge metabolic turnover and energetic demands. The vast majority of its energy is produced and cycled in form of ATP which stems mainly from oxidative phosphorylation occurring at the respiratory chain in the mitochondria. Apart from energy production, the respiratory chain is also the main source of reactive oxygen species and plays a pivotal role in the regulation of oxidative stress. Dysfunction of the respiratory chain is therefore found in most common heart conditions. The pathophysiology of mitochondrial respiratory chain dysfunction in hereditary cardiac mitochondrial disease, the ageing heart, in LV hypertrophy and heart failure, and in ischaemia-reperfusion injury is reviewed. We introduce the practising clinician to the complex physiology of the respiratory chain, highlight its impact on common cardiac disorders and review translational pharmacological and non-pharmacological treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Christopher J Neil
- University of Aberdeen, United Kingdom; Western Health, Victoria, Australia
| | | | | |
Collapse
|
37
|
Hiemstra JA, Liu S, Ahlman MA, Schuleri KH, Lardo AC, Baines CP, Dellsperger KC, Bluemke DA, Emter CA. A new twist on an old idea: a two-dimensional speckle tracking assessment of cyclosporine as a therapeutic alternative for heart failure with preserved ejection fraction. Physiol Rep 2013; 1:e00174. [PMID: 24744855 PMCID: PMC3970750 DOI: 10.1002/phy2.174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/01/2013] [Accepted: 11/04/2013] [Indexed: 01/12/2023] Open
Abstract
We recently reported that mitochondrial dysfunction, characterized by increased mitochondrial permeability transition (MPT), was present in a translational swine model of heart failure with preserved ejection fraction (HFpEF). Cyclophilin D is a key component of the MPT pore, therefore, the purpose of this study was to test the efficacy of a novel cyclosporine (CsA) dosing scheme as a therapeutic alternative for HFpEF. Computed tomography (CT), two‐dimensional speckle tracking two‐dimensional speckle tracking (2DST), and invasive hemodynamics were used to evaluate cardiac function. CT imaging showed 14 weeks of CsA treatment caused eccentric myocardial remodeling (contrasting concentric remodeling in untreated HF animals) and elevated systemic pressures. 2DST detected left ventricular (LV) mechanics associated with systolic and diastolic dysfunction prior to the onset of significantly increased LV end diastolic pressure including: (1) decreased systolic apical rotation rate, longitudinal displacement, and longitudinal/radial/circumferential strain; (2) decreased early diastolic untwisting and longitudinal strain rate; and (3) increased late diastolic radial/circumferential mitral strain rate. LV mechanics associated with systolic and diastolic impairment was enhanced to a greater extent than seen in untreated HF animals following CsA treatment. In conclusion, CsA treatment accelerated the development of heart failure, including dilatory LV remodeling and impaired systolic and diastolic mechanics. Although our findings do not support CsA as a viable therapy for HFpEF, 2DST was effective in differentiating between progressive gradations of developing HF and detecting diastolic impairment prior to the development of overt diastolic dysfunction. We recently reported that mitochondrial dysfunction, characterized by increased mitochondrial permeability transition (MPT), was present in a translational swine model of heart failure with preserved ejection fraction (HFpEF). Cyclophilin D is a key component of the MPT pore, therefore, the purpose of this study was to test the efficacy of a novel cyclosporine (CsA) dosing scheme as a therapeutic alternative for HFpEF. CsA treatment accelerated the development of heart failure, including dilatory LV remodeling and impaired systolic and diastolic mechanics. Although our findings do not support CsA as a viable therapy for HFpEF, 2DST was effective in differentiating between progressive gradations of developing HF and detecting diastolic impairment prior to the development of overt diastolic dysfunction.
Collapse
Affiliation(s)
- Jessica A Hiemstra
- Department of Biomedical Science, University of Missouri- Columbia, Columbia, Missouri
| | - Songtao Liu
- Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Maryland ; Molecular Biomedical Imaging Laboratory, National Institute of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| | - Mark A Ahlman
- Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Maryland ; Molecular Biomedical Imaging Laboratory, National Institute of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| | - Karl H Schuleri
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Albert C Lardo
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Christopher P Baines
- Department of Biomedical Science, University of Missouri- Columbia, Columbia, Missouri ; Dalton Cardiovascular Research Center, University of Missouri- Columbia, Columbia, Missouri
| | - Kevin C Dellsperger
- Department of Medical Pharmacology and Physiology, University of Missouri- Columbia, Columbia, Missouri ; Department of Internal Medicine, University of Missouri- Columbia, Columbia, Missouri ; Center for Health Care Quality, University of Missouri- Columbia, 1600 E. RollinsW160 Veterinary Medicine, Columbia, 65211, Missouri
| | - David A Bluemke
- Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Maryland ; Molecular Biomedical Imaging Laboratory, National Institute of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| | - Craig A Emter
- Department of Biomedical Science, University of Missouri- Columbia, Columbia, Missouri
| |
Collapse
|
38
|
Khan RS, Lin Y, Hu Y, Son NH, Bharadwaj KG, Palacios C, Chokshi A, Ji R, Yu S, Homma S, Schulze PC, Tian R, Goldberg IJ. Rescue of heart lipoprotein lipase-knockout mice confirms a role for triglyceride in optimal heart metabolism and function. Am J Physiol Endocrinol Metab 2013; 305:E1339-47. [PMID: 24085031 PMCID: PMC3882371 DOI: 10.1152/ajpendo.00349.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hearts utilize fatty acids as a primary source of energy. The sources of those lipids include free fatty acids and lipoprotein triglycerides. Deletion of the primary triglyceride-hydrolyzing enzyme lipoprotein lipase (LPL) leads to cardiac dysfunction. Whether heart LPL-knockout (hLPL0) mice are compromised due a deficiency in energetic substrates is unknown. To test whether alternative sources of energy will prevent cardiac dysfunction in hLPL0 mice, two different models were used to supply nonlipid energy. 1) hLPL0 mice were crossed with mice transgenically expressing GLUT1 in cardiomyocytes to increase glucose uptake into the heart; this cross-corrected cardiac dysfunction, reduced cardiac hypertrophy, and increased myocardial ATP. 2) Mice were randomly assigned to a sedentary or training group (swimming) at 3 mo of age, which leads to increased skeletal muscle production of lactate. hLPL0 mice had greater expression of the lactate transporter monocarboxylate transporter-1 (MCT-1) and increased cardiac lactate uptake. Compared with hearts from sedentary hLPL0 mice, hearts from trained hLPL0 mice had adaptive hypertrophy and improved cardiac function. We conclude that defective energy intake and not the reduced uptake of fat-soluble vitamins or cholesterol is responsible for cardiac dysfunction in hLPL0 mice. In addition, our studies suggest that adaptations in cardiac metabolism contribute to the beneficial effects of exercise on the myocardium of patients with heart failure.
Collapse
|
39
|
Kraljevic J, Marinovic J, Pravdic D, Zubin P, Dujic Z, Wisloff U, Ljubkovic M. Aerobic interval training attenuates remodelling and mitochondrial dysfunction in the post-infarction failing rat heart. Cardiovasc Res 2013; 99:55-64. [PMID: 23554460 DOI: 10.1093/cvr/cvt080] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Following a large myocardial infarction (MI), remaining viable muscle often undergoes pathological remodelling and progresses towards chronic heart failure. Mitochondria may also be affected by this process and, due to their functional importance, likely contribute to the progression of the disease. Aerobic interval training (AIT) has been shown effective in diminishing pathological myocardial transformation, but the effects of AIT on mitochondrial function in hearts undergoing remodelling are not known. METHODS AND RESULTS Adult female Sprague-Dawley rats were randomized to either 8 weeks of aerobic interval treadmill running (5 days/week), which started 4 weeks after left coronary artery ligation (MI-Trained), or a sedentary group (MI-Sedentary). Echocardiography was performed before and after the 8-week period, at which point the left ventricles (LVs) were also harvested. Twelve weeks after surgery, MI-Sedentary rats had significantly lower LV fractional shortening compared with MI-Trained rats. Complex I-dependent respiration assessed in isolated LV mitochondria was decreased by ∼37% in MI-Sedentary and 17% in MI-Trained animals (group differences P < 0.05), compared with sham-operated animals. This was paralleled with diminished ATP production and increased degree of protein oxidation in MI-Sedentary rats. The enzymatic activity of complex I was also decreased to a greater extent in MI-Sedentary than in MI-Trained animals, with no evidence of its reduced expression. When complex II substrate was used, no differences among the three groups were observed. CONCLUSION Exercise reduces LV contractile deterioration in post-infarction heart failure and alleviates the extent of mitochondrial dysfunction, which is paralleled with preserved complex I activity.
Collapse
Affiliation(s)
- Jasenka Kraljevic
- Department of Physiology, University of Split School of Medicine, Soltanska 2, Split 21000, Croatia
| | | | | | | | | | | | | |
Collapse
|
40
|
Effects of local mechanical stimulation on coronary artery endothelial function and angiotensin II type 1 receptor in pressure or flow-overload. J Hypertens 2013; 31:720-9. [DOI: 10.1097/hjh.0b013e32835d6d2c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Marshall KD, Muller BN, Krenz M, Hanft LM, McDonald KS, Dellsperger KC, Emter CA. Heart failure with preserved ejection fraction: chronic low-intensity interval exercise training preserves myocardial O2 balance and diastolic function. J Appl Physiol (1985) 2013; 114:131-47. [PMID: 23104696 PMCID: PMC3544520 DOI: 10.1152/japplphysiol.01059.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/23/2012] [Indexed: 12/17/2022] Open
Abstract
We have previously reported chronic low-intensity interval exercise training attenuates fibrosis, impaired cardiac mitochondrial function, and coronary vascular dysfunction in miniature swine with left ventricular (LV) hypertrophy (Emter CA, Baines CP. Am J Physiol Heart Circ Physiol 299: H1348-H1356, 2010; Emter CA, et al. Am J Physiol Heart Circ Physiol 301: H1687-H1694, 2011). The purpose of this study was to test two hypotheses: 1) chronic low-intensity interval training preserves normal myocardial oxygen supply/demand balance; and 2) training-dependent attenuation of LV fibrotic remodeling improves diastolic function in aortic-banded sedentary, exercise-trained (HF-TR), and control sedentary male Yucatan miniature swine displaying symptoms of heart failure with preserved ejection fraction. Pressure-volume loops, coronary blood flow, and two-dimensional speckle tracking ultrasound were utilized in vivo under conditions of increasing peripheral mean arterial pressure and β-adrenergic stimulation 6 mo postsurgery to evaluate cardiac function. Normal diastolic function in HF-TR animals was characterized by prevention of increased time constant of isovolumic relaxation, normal LV untwisting rate, and enhanced apical circumferential and radial strain rate. Reduced fibrosis, normal matrix metalloproteinase-2 and tissue inhibitors of metalloproteinase-4 mRNA expression, and increased collagen III isoform mRNA levels (P < 0.05) accompanied improved diastolic function following chronic training. Exercise-dependent improvements in coronary blood flow for a given myocardial oxygen consumption (P < 0.05) and cardiac efficiency (stroke work to myocardial oxygen consumption, P < 0.05) were associated with preserved contractile reserve. LV hypertrophy in HF-TR animals was associated with increased activation of Akt and preservation of activated JNK/SAPK. In conclusion, chronic low-intensity interval exercise training attenuates diastolic impairment by promoting compliant extracellular matrix fibrotic components and preserving extracellular matrix regulatory mechanisms, preserves myocardial oxygen balance, and promotes a physiological molecular hypertrophic signaling phenotype in a large animal model resembling heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Kurt D Marshall
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri 65211, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen YM, Li ZB, Zhu M, Cao YM. Effects of exercise training on left ventricular remodelling in heart failure patients: an updated meta-analysis of randomised controlled trials. Int J Clin Pract 2012; 66:782-791. [PMID: 22805270 DOI: 10.1111/j.1742-1241.2012.02942.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objectives: The purpose of this updated meta-analysis was to determine whether exercise training reversed left ventricular remodelling in heart failure patients. Methods: Articles were searched in MEDLINE, Cochrane Central Register of Controlled Trials, CINAHL, EMBASE and PubMed (up until October, 2011). The anti-remodelling benefit of exercise training was assessed by ejection fraction (EF), end-diastolic volume (EDV) and end-systolic volume (ESV). Standardised mean difference (SMD) together with 95% confidence intervals (CI) was calculated. Subgroup meta-analysis with exercise duration was also conducted. Results: Fifteen randomised controlled trials with 813 patients were included. Aerobic exercise training improved EF (SMD = 0.44; 95% CI 0.28 to 0.61), EDV (SMD = -0.33; 95% CI -0.49 to -0.16) and ESV (SMD = -0.40; 95% CI -0.57 to -0.23). Subgroup analysis indicated that long-term aerobic exercise (≥6 months) had a marked positive effect on EF (SMD = 0.5; 95% confidence interval 0.31 to 0.69), EDV (SMD = -0.38; 95% CI -0.57 to -0.19) and ESV (SMD = -0.48; 95% CI -0.67 to -0.29), but there was no evidence of benefit with short-term aerobic exercise (<6 months): EF (SMD = 0.27; 95% CI -0.08 to 0.61), EDV (SMD = -0.14; 95% CI -0.48 to 0.21) and ESV (SMD = -0.08; 95% CI -0.47 to 0.30). Strength training (alone or plus aerobic training) was not associated with improvements in EDV and ESV, with all confidence intervals including 0. Conclusions: Aerobic exercise training, especially long-term duration (≥6 months) reverses left ventricular remodelling in clinically stable patients with heart failure. Strength training (alone or plus aerobic training) did not improve or worsen ventricular remodelling.
Collapse
Affiliation(s)
- Y M Chen
- Division of Geriatric Cardiology, General Hospital of Chinese PLA, Beijing, China
| | | | | | | |
Collapse
|
43
|
McDonald KS, Hanft LM, Domeier TL, Emter CA. Length and PKA Dependence of Force Generation and Loaded Shortening in Porcine Cardiac Myocytes. Biochem Res Int 2012; 2012:371415. [PMID: 22844597 PMCID: PMC3398585 DOI: 10.1155/2012/371415] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/01/2012] [Indexed: 11/17/2022] Open
Abstract
In healthy hearts, ventricular ejection is determined by three myofibrillar properties; force, force development rate, and rate of loaded shortening (i.e., power). The sarcomere length and PKA dependence of these mechanical properties were measured in porcine cardiac myocytes. Permeabilized myocytes were prepared from left ventricular free walls and myocyte preparations were calcium activated to yield ~50% maximal force after which isometric force was measured at varied sarcomere lengths. Porcine myocyte preparations exhibited two populations of length-tension relationships, one being shallower than the other. Moreover, myocytes with shallow length-tension relationships displayed steeper relationships following PKA. Sarcomere length-K(tr) relationships also were measured and K(tr) remained nearly constant over ~2.30 μm to ~1.90 μm and then increased at lengths below 1.90 μm. Loaded-shortening and peak-normalized power output was similar at ~2.30 μm and ~1.90 μm even during activations with the same [Ca(2+)], implicating a myofibrillar mechanism that sustains myocyte power at lower preloads. PKA increased myocyte power and yielded greater shortening-induced cooperative deactivation in myocytes, which likely provides a myofibrillar mechanism to assist ventricular relaxation. Overall, the bimodal distribution of myocyte length-tension relationships and the PKA-mediated changes in myocyte length-tension and power are likely important modulators of Frank-Starling relationships in mammalian hearts.
Collapse
Affiliation(s)
- Kerry S. McDonald
- Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Laurin M. Hanft
- Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Timothy L. Domeier
- Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Craig A. Emter
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
44
|
McCommis KS, McGee AM, Laughlin MH, Bowles DK, Baines CP. Hypercholesterolemia increases mitochondrial oxidative stress and enhances the MPT response in the porcine myocardium: beneficial effects of chronic exercise. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1250-8. [PMID: 21865543 PMCID: PMC3213933 DOI: 10.1152/ajpregu.00841.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 08/18/2011] [Indexed: 02/06/2023]
Abstract
Hypercholesterolemia has been suggested to have direct negative effects on myocardial function due to increased reactive oxygen species (ROS) generation and increased myocyte death. Mitochondrial permeability transition (MPT) is a significant mediator of cell death, which is enhanced by ROS generation and attenuated by exercise training. The purpose of this study was to investigate the effect of hypercholesterolemia on the MPT response of cardiac mitochondria. We tested the hypothesis that familial hypercholesterolemic (FH) pigs would have an enhanced MPT response and that exercise training could reverse this phenotype. MPT was assessed by mitochondrial swelling in response to 10-100 μM Ca(2+). FH pigs did show an increased MPT response to Ca(2+) that was associated with decreases in the expression of the putative MPT pore components mitochondrial phosphate carrier (PiC) and cyclophilin-D (CypD). FH also caused increased oxidative stress, depicted by increased protein nitrotyrosylation, as well as decreased levels of reduced GSH in cardiac mitochondria. Expression of the mitochondrial antioxidant enzymes manganese superoxide dismutase (MnSOD), thioredoxin-2 (Trx2), and peroxiredoxin-3 (Prx3) was greatly reduced in the FH pigs. In contrast, cytosolic catalase expression and activity were increased. However, chronic exercise training was able to normalize the MPT response in FH pigs, reduce mitochondrial oxidative stress, and return MnSOD, Trx2, Prx3, and catalase expression/activities to normal. We conclude that FH reduces mitochondrial antioxidants, increases mitochondrial oxidative stress, and enhances the MPT response in the porcine myocardium, and that exercise training can reverse these detrimental alterations.
Collapse
Affiliation(s)
- Kyle S McCommis
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri 65211, USA
| | | | | | | | | |
Collapse
|
45
|
Emter CA, Tharp DL, Ivey JR, Ganjam VK, Bowles DK. Low-intensity interval exercise training attenuates coronary vascular dysfunction and preserves Ca²⁺-sensitive K⁺ current in miniature swine with LV hypertrophy. Am J Physiol Heart Circ Physiol 2011; 301:H1687-94. [PMID: 21841018 DOI: 10.1152/ajpheart.00610.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Coronary vascular dysfunction has been observed in several models of heart failure (HF). Recent evidence indicates that exercise training is beneficial for patients with HF, but the precise intensity and underlying mechanisms are unknown. Left ventricular (LV) hypertrophy can play a significant role in the development of HF; therefore, the purpose of this study was to assess the effects of low-intensity interval exercise training on coronary vascular function in sedentary (HF) and exercise trained (HF-TR) aortic-banded miniature swine displaying LV hypertrophy. Six months postsurgery, in vivo coronary vascular responses to endothelin-1 (ET-1) and adenosine were measured in the left anterior descending coronary artery. Baseline and maximal coronary vascular conductance were similar between all groups. ET-1-induced reductions in coronary vascular conductance (P < 0.05) were greater in HF vs. sedentary control and HF-TR groups. Pretreatment with the ET type A (ET(A)) receptor blocker BQ-123 prevented ET-1 hypersensitivity in HF animals. Whole cell voltage clamp was used to characterize composite K(+) currents (I(K(+))) in coronary smooth muscle cells. Raising internal Ca(2+) from 200 to 500 nM increased Ca(2+)-sensitive K(+) current in HF-TR and control, but not HF animals. In conclusion, an ET(A)-receptor-mediated hypersensitivity to ET-1, elevated resting LV wall tension, and decreased coronary smooth muscle cell Ca(2+)-sensitive I(K(+)) was found in sedentary animals with LV hypertrophy. Low-intensity interval exercise training preserved normal coronary vascular function and smooth muscle cell Ca(2+)-sensitive I(K(+)), illustrating a potential mechanism underlying coronary vascular dysfunction in a large-animal model of LV hypertrophy. Our results demonstrate the potential clinical impact of exercise on coronary vascular function in HF patients displaying pathological LV hypertrophy.
Collapse
Affiliation(s)
- Craig A Emter
- Dept. of Biomedical Science, Univ. of Missouri-Columbia, 1600 E. Rollins, E117 Veterinary Medicine, Columbia, MO 65211, USA.
| | | | | | | | | |
Collapse
|