1
|
Diaz Sanmartin LA, Gruslova AB, Nolen DR, Feldman MD, Valvano JW. Measurement of left ventricular volume with admittance incorporated onto percutaneous ventricular assist device. Med Biol Eng Comput 2024:10.1007/s11517-024-03168-y. [PMID: 39004652 DOI: 10.1007/s11517-024-03168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
Percutaneous ventricular assist devices (pVADs) incorporated with admittance electrodes have been validated in animal studies for accurate instantaneous volumetric measurements. Since miniaturization of the pVAD profile is a priority to reduce vascular complications in patients, our study aimed to validate admittance measurements using three electrodes instead of the standard four. Complex admittance was measured between an electrode pair and a pVAD metallic blood-intake tip, both with finite element analysis and on the benchtop. The catheter and electrode arrays were first simulated inside prolate ellipsoid models of the left ventricle (LV) demonstrating current flow throughout all parts of the LV as well as minimal influence of off-center catheter placement in the recorded signal. Admittance measurements were validated in 3D-printed models of healthy and dilated hearts (100-400 mL end-diastolic volumes). Minimal interference between a pVAD motor and the current signal of our admittance system was demonstrated. A modified Wei's equation focused on three electrodes was developed to be compatible with reduced profile pVADs occurring clinically, incorporated with admittance electrodes and wires. The modified equation was compared against Wei's original equation showing improved accuracy of calculated volumes. Reducing electrode footprint can simplify the incorporation of Admittance technology on any pVAD, allowing for instantaneous recognition of native heart recovery and assistance with pVAD weaning.
Collapse
Affiliation(s)
- Luis A Diaz Sanmartin
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aleksandra B Gruslova
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Drew R Nolen
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marc D Feldman
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jonathan W Valvano
- Department of Electrical Engineering, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
2
|
Aslam MI, Gruslova AB, Almomani A, Nolen D, Elliott JJ, Jani VP, Kottam A, Porterfield J, Heighten C, Anderson AS, Valvano JW, Feldman MD. Modification of a Transvalvular Microaxial Flow Pump for Instantaneous Determination of Native Cardiac Output and Volume. J Card Fail 2023; 29:1369-1379. [PMID: 37105397 DOI: 10.1016/j.cardfail.2023.04.007] [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: 11/21/2022] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND The current Impella cardiopulmonary (CP) pump, used for mechanical circulatory support in patients with cardiogenic shock (CS), cannot assess native cardiac output (CO) and left ventricular (LV) volumes. These data are valuable in facilitating device management and weaning. Admittance technology allows for accurate assessment of cardiac chamber volumes. OBJECTIVES This study tested the ability to engineer admittance electrodes onto an existing Impella CP pump to assess total and native CO as well as LV chamber volumes in an instantaneous manner. METHODS Impella CP pumps were fitted with 4 admittance electrodes and were placed in the LVs of adult swine (n = 9) that were subjected to 3 different hemodynamic conditions, including Impella CP speed adjustments, administration of escalating doses of dobutamine and microsphere injections into the left main artery to result in cardiac injury. CO, according to admittance electrodes, was calculated from LV volumes and heart rate. In addition, CO was calculated in each instance via thermodilution, continuous CO measurement, the Fick principle, and aortic velocity-time integral by means of echocardiography. RESULTS Modified Impella CP pumps were placed in swine LVs successfully. CO, as determined by admittance electrodes, was similar by trend to other methods of CO assessment. It was corrected for pump speed to calculate native CO, and calculated LV chamber volumes trended as expected in each experimental protocol. CONCLUSIONS We report, for the first time, that an Impella CP pump can be fitted with admittance electrodes and used to determine total and native CO in various hemodynamic situations. CONDENSED ABSTRACT Transvalvular mechanical circulatory support devices such as the Impella CP do not have the ability to provide real-time information on native cardiac output (CO) and left ventricular (LV) volumes. This information is critical in device management and in weaning in patients with cardiogenic shock. We demonstrate, for the first time, that Impella CP pumps coupled with admittance electrodes are able to determine native CO and LV chamber volumes in multiple hemodynamic situations such as Impella pump speed adjustments, escalating dobutamine administration and cardiac injury from microsphere injection.
Collapse
Affiliation(s)
- M Imran Aslam
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Aleksandra B Gruslova
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ahmed Almomani
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Drew Nolen
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - James J Elliott
- Department of Laboratory Animal Resources, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Vivek P Jani
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anil Kottam
- BridgeSource Medical Corporation, Austin, Texas
| | | | | | - Allen S Anderson
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jonathan W Valvano
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas
| | - Marc D Feldman
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas.
| |
Collapse
|
3
|
Bowers SLK, Meng Q, Kuwabara Y, Huo J, Minerath R, York AJ, Sargent MA, Prasad V, Saviola AJ, Galindo DC, Hansen KC, Vagnozzi RJ, Yutzey KE, Molkentin JD. Col1a2-Deleted Mice Have Defective Type I Collagen and Secondary Reactive Cardiac Fibrosis with Altered Hypertrophic Dynamics. Cells 2023; 12:2174. [PMID: 37681905 PMCID: PMC10486458 DOI: 10.3390/cells12172174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
RATIONALE The adult cardiac extracellular matrix (ECM) is largely comprised of type I collagen. In addition to serving as the primary structural support component of the cardiac ECM, type I collagen also provides an organizational platform for other ECM proteins, matricellular proteins, and signaling components that impact cellular stress sensing in vivo. OBJECTIVE Here we investigated how the content and integrity of type I collagen affect cardiac structure function and response to injury. METHODS AND RESULTS We generated and characterized Col1a2-/- mice using standard gene targeting. Col1a2-/- mice were viable, although by young adulthood their hearts showed alterations in ECM mechanical properties, as well as an unanticipated activation of cardiac fibroblasts and induction of a progressive fibrotic response. This included augmented TGFβ activity, increases in fibroblast number, and progressive cardiac hypertrophy, with reduced functional performance by 9 months of age. Col1a2-loxP-targeted mice were also generated and crossed with the tamoxifen-inducible Postn-MerCreMer mice to delete the Col1a2 gene in myofibroblasts with pressure overload injury. Interestingly, while germline Col1a2-/- mice showed gradual pathologic hypertrophy and fibrosis with aging, the acute deletion of Col1a2 from activated adult myofibroblasts showed a loss of total collagen deposition with acute cardiac injury and an acute reduction in pressure overload-induce cardiac hypertrophy. However, this reduction in hypertrophy due to myofibroblast-specific Col1a2 deletion was lost after 2 and 6 weeks of pressure overload, as fibrotic deposition accumulated. CONCLUSIONS Defective type I collagen in the heart alters the structural integrity of the ECM and leads to cardiomyopathy in adulthood, with fibroblast expansion, activation, and alternate fibrotic ECM deposition. However, acute inhibition of type I collagen production can have an anti-fibrotic and anti-hypertrophic effect.
Collapse
Affiliation(s)
- Stephanie L. K. Bowers
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Qinghang Meng
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
- Center for Organoid and Regeneration Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou 511466, China
| | - Yasuhide Kuwabara
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jiuzhou Huo
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Rachel Minerath
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Allen J. York
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michelle A. Sargent
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Vikram Prasad
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Anthony J. Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Ceja Galindo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ronald J. Vagnozzi
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Cardiology, Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jeffery D. Molkentin
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
4
|
Florens N, Kasam RK, Rudman-Melnick V, Lin SC, Prasad V, Molkentin JD. Interleukin-33 Mediates Cardiomyopathy After Acute Kidney Injury by Signaling to Cardiomyocytes. Circulation 2023; 147:746-758. [PMID: 36695175 PMCID: PMC9992318 DOI: 10.1161/circulationaha.122.063014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/19/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is a short-term life-threatening condition that, if survived, can lead to renal insufficiency and development of chronic kidney disease. The pathogenesis of AKI and chronic kidney disease involves direct effects on the heart and the development of hypertrophy and cardiomyopathy. METHODS We used mouse models of ischemia/reperfusion AKI and unilateral ureteral obstruction to investigate the role of IL-33 (interleukin-33) and its receptor-encoding gene Il1rl1 (also called ST2L [suppression of tumorigenicity 2]) in cardiac remodeling after AKI. Mice with cell type-specific genetic disruption of the IL-33/ST2L axis were used, and IL-33 monoclonal antibody, adeno-associated virus encoding IL-33 or ST2L, and recombinant IL-33, as well. RESULTS Mice deficient in Il33 were refractory to cardiomyopathy associated with 2 models of kidney injury. Treatment of mice with monoclonal IL-33 antibody also protected the heart after AKI. Moreover, overexpression of IL-33 or injection of recombinant IL-33 induced cardiac hypertrophy or cardiomyopathy, but not in mice lacking Il1rl1. AKI-induced cardiomyopathy was also reduced in mice with cardiac myocyte-specific deletion of Il1rl1 but not in endothelial cell- or fibroblast-specific deletion of Il1rl1. Last, overexpression of the ST2L receptor in cardiac myocytes recapitulated induction of cardiac hypertrophy. CONCLUSIONS These results indicate that IL-33 released from the kidney during AKI underlies cardiorenal syndrome by directly signaling to cardiac myocytes, suggesting that antagonism of IL-33/ST2 axis would be cardioprotective in patients with kidney disease.
Collapse
Affiliation(s)
- Nans Florens
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Rajesh K. Kasam
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Valeria Rudman-Melnick
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Suh-Chin Lin
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D. Molkentin
- Department of Pediatrics, Cincinnati Children’s Hospital and the University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
5
|
Gruslova AB, Cabe AG, Kottam A, Walmsley J, Porterfield JE, Sako EY, Feldman MD, Valvano JW. Smart Drain for Post-Cardiac Surgery Left Ventricular Volumes Evaluated in Large Animal Models. Ann Thorac Surg 2022; 114:2270-2279. [PMID: 34890574 PMCID: PMC9170842 DOI: 10.1016/j.athoracsur.2021.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/17/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Open heart surgeries for coronary arterial bypass graft and valve replacements are performed on 400,000 Americans each year. Unexplained hypotension during recovery causes morbidity and mortality through cerebral, kidney, and coronary hypoperfusion. An early detection method that distinguishes between hypovolemia and decreased myocardial function before onset of hypotension is desirable. We hypothesized that admittance measured from a modified pericardial drain can detect changes in left ventricular end-systolic, end-diastolic, and stroke volumes. METHODS Admittance was measured from 2 modified pericardial drains placed in 7 adult female dogs using an open chest preparation, each with 8 electrodes. The resistive and capacitive components of the measured admittance signal were used to distinguish blood and muscle components. Admittance measurements were taken from 12 electrode configurations in each experiment. Left ventricular preload was reduced by inferior vena cava occlusion. Physiologic response to vena cava occlusion was measured by aortic pressure, aortic flow, left ventricle diameter, left ventricular wall thickness, and electrocardiogram. RESULTS Admittance successfully detected a drop in left ventricular end-diastolic volume (P < .001), end-systolic volume (P < .001), and stroke volume (P < .001). Measured left ventricular muscle resistance correlated with crystal-derived left ventricular wall thickness (R2 = 0.96), validating the method's ability to distinguish blood from muscle components. CONCLUSIONS Admittance measured from chest tubes can detect changes in left ventricular end-systolic, end-diastolic, and stroke volumes and may therefore have diagnostic value for unexplained hypotension.
Collapse
Affiliation(s)
- Aleksandra B Gruslova
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Andrew G Cabe
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | | | - John Walmsley
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas
| | | | - Edward Y Sako
- Department of Cardiothoracic Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Marc D Feldman
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jonathan W Valvano
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas.
| |
Collapse
|
6
|
Miranda-Silva D, Sequeira V, Lourenço AP, Falcão-Pires I. Assessing Rodent Cardiac Function in vivo Using Hemodynamic Pressure-Volume Loops. Front Physiol 2022; 12:751326. [PMID: 35811573 PMCID: PMC9260141 DOI: 10.3389/fphys.2021.751326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) triggered by cardiovascular and non-cardiovascular diseases is a leading cause of death worldwide and translational research is urgently needed to better understand the mechanisms of the failing heart. For this purpose, rodent models of heart disease combined with in vivo cardiac functional assessment have provided valuable insights into the physiological significance of a given genetic or pharmacological modification. In small animals, cardiac function and structure can be evaluated by methods such as echocardiography, telemetry or hemodynamics using conductance catheters. Indeed, hemodynamic analysis of pressure-volume loops (PV-loops) has become the gold standard methodology to study in vivo cardiac function in detail. This method provides simultaneous measurement of both pressure and volume signals from rodents intact beating hearts. On the one hand, PV-loop analysis has deeply expanded the knowledge on molecular cardiac physiology by allowing establishing important functional correlations. On the other hand, these measurements allow dissecting the cardiovascular functional impact of certain therapeutic interventions or specific signaling pathways using transgenic models of disease. However, a detailed assessment of cardiac function and structure in vivo still warrants proper standardization and optimization to boost the progress of HF research. With increasing concerns over data accuracy and reproducibility, guidelines and best practices for cardiac physiology measurements in experimental settings are needed. This article aims to review the best practices for carrying out cardiac hemodynamic assessment using PV-loops in vivo in rodents intact beating hearts, also providing an overview of its advantages, disadvantages and applications in cardiovascular research.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Vasco Sequeira
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Translational Science, DZHI, Universitätsklinikum Würzburg, Würzburg, Germany
| | - André P. Lourenço
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- *Correspondence: Inês Falcão-Pires,
| |
Collapse
|
7
|
Fukami H, Sunada K. Effects of vasopressin administration in the oral cavity on cardiac function and hemodynamics in rats. J Dent Anesth Pain Med 2022; 22:11-18. [PMID: 35169616 PMCID: PMC8814726 DOI: 10.17245/jdapm.2022.22.1.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/21/2021] [Accepted: 12/20/2021] [Indexed: 11/15/2022] Open
Abstract
Background The vasoconstrictive effect of epinephrine in local anesthetics affects the heart, which leads to hesitation among dentists in injecting local anesthetics into patients with cardiovascular disease. Due to its vasoconstrictive effects, the present study investigated the effects of vasopressin administration on cardiac function in rats. Methods Experiment 1 aimed to determine the vasopressin concentration that could affect cardiac function. An arterial catheter was inserted into the male Wistar rats. Next, 0.03, 0.3, and 3.0 U/mL arginine vasopressin (AVP) (0.03V, 0.3V, and 3.0V) was injected into the tongue, and the blood pressure was measured. The control group received normal saline only. In Experiment 2, following anesthesia infiltration, a pressure–volume catheter was placed in the left ventricle. Baseline values of end-systolic elastance, end-diastolic volume, end-systolic pressure, stroke work, stroke volume, and end-systolic elastance were recorded. Next, normal saline and 3.0V AVP were injected into the tongue to measure their effect on hemodynamic and cardiac function. Results After 3.0V administration, systolic blood pressures at 10 and 15 min were higher than those of the control group; they increased at 10 min compared with those at baseline. The diastolic blood pressures at 5–15 min were higher than those of the control group; they increased at 5 and 10 min compared with those at baseline. The preload decreased at 5 and 10 min compared to that at baseline. However, the afterload increased from 5 to 15 min compared with that of the control group; it increased at 10 min compared with that at baseline. Stroke volume decreased at 10 and 15 min compared with that of the control group; it decreased from 5 to 15 min compared with that at baseline. Stroke work decreased from 5 to 15 min compared with that of the control group; it decreased from 5 to 15 min compared with that at baseline. Conclusion Our results showed that 3.0 U/mL concentration of vasopressin resulted in increased blood pressure, decreased stroke volume and stoke work, decreased preload and increased afterload, without any effect on myocardial contractility.
Collapse
Affiliation(s)
- Hayato Fukami
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Hansen ESS, Madsen TL, Wood G, Granfeldt A, Bøgh N, Tofig BJ, Agger P, Lindhardt JL, Poulsen CB, Bøtker HE, Kim WY. Veno-occlusive unloading of the heart reduces infarct size in experimental ischemia-reperfusion. Sci Rep 2021; 11:4483. [PMID: 33627745 PMCID: PMC7904802 DOI: 10.1038/s41598-021-84025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022] Open
Abstract
Mechanical unloading of the left ventricle reduces infarct size after acute myocardial infarction by reducing cardiac work. Left ventricular veno-occlusive unloading reduces cardiac work and may reduce ischemia and reperfusion injury. In a porcine model of myocardial ischemia-reperfusion injury we randomized 18 pigs to either control or veno-occlusive unloading using a balloon engaged from the femoral vein into the inferior caval vein and inflated at onset of ischemia. Evans blue and 2,3,5-triphenyltetrazolium chloride were used to determine the myocardial area at risk and infarct size, respectively. Pressure-volume loops were recorded to calculate cardiac work, left ventricular (LV) volumes and ejection fraction. Veno-occlusive unloading reduced infarct size compared with controls (Unloading 13.9 ± 8.2% versus Control 22.4 ± 6.6%; p = 0.04). Unloading increased myocardial salvage (54.8 ± 23.4% vs 28.5 ± 14.0%; p = 0.02), while the area at risk was similar (28.4 ± 6.7% vs 27.4 ± 5.8%; p = 0.74). LV ejection fraction was preserved in the unloaded group, while the control group showed a reduced LV ejection fraction. Veno-occlusive unloading reduced myocardial infarct size and preserved LV ejection fraction in an experimental acute ischemia-reperfusion model. This proof-of-concept study demonstrated the potential of veno-occlusive unloading as an adjunctive cardioprotective therapy in patients undergoing revascularization for acute myocardial infarction.
Collapse
Affiliation(s)
- Esben Søvsø Szocska Hansen
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard, 8200, Aarhus N, Denmark
| | - Tobias Lynge Madsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Gregory Wood
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Asger Granfeldt
- Department of Intensive Care Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 8200, Aarhus N, Denmark
| | - Nikolaj Bøgh
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard, 8200, Aarhus N, Denmark
| | - Bawer Jalal Tofig
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Peter Agger
- Department of Clinical Medicine, Comparative Medicine Lab, Aarhus University, Palle Juul-Jensens Boulevard, 8200, Aarhus N, Denmark
| | - Jakob Lykke Lindhardt
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Christian Bo Poulsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark. .,Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard, 8200, Aarhus N, Denmark.
| |
Collapse
|
9
|
Kambis TN, Shahshahan HR, Kar S, Yadav SK, Mishra PK. Transgenic Expression of miR-133a in the Diabetic Akita Heart Prevents Cardiac Remodeling and Cardiomyopathy. Front Cardiovasc Med 2019; 6:45. [PMID: 31069235 PMCID: PMC6491745 DOI: 10.3389/fcvm.2019.00045] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/25/2019] [Indexed: 01/22/2023] Open
Abstract
Advanced diabetes mellitus (DM) may have both insulin resistance and deficiency (double DM) that accelerates diabetic cardiomyopathy (DMCM), a cardiac muscle disorder. Reduced cardiac miR-133a, a cardioprotective miRNA, is associated with DMCM. However, it is unclear whether increasing miR-133a levels in the double DM heart could prevent DMCM. We hypothesized that increasing cardiac levels of miR-133a could prevent DMCM in Akita, a mouse model of double DM. To test the hypothesis, we created Akita/miR-133aTg mice, a new strain of Akita where miR-133a is overexpressed in the heart, by crossbreeding male Akita with female cardiac-specific miR-133a transgenic mice. We validated Akita/miR-133aTg mice by genotyping and phenotyping (miR-133a levels in the heart). To determine whether miR-133a overexpression could prevent cardiac remodeling and cardiomyopathy, we evaluated cardiac fibrosis, hypertrophy, and dysfunction (P-V loop) in 13-15 week male WT, Akita, Akita/miR-133aTg, and miR-133aTg mice. Our results revealed that miR-133a overexpression in the Akita heart prevents DM-induced cardiac fibrosis (reduced collagen deposition), hypertrophy (decreased beta-myosin heavy chain), and impaired contractility (downregulated calcium handling protein sarco-endoplasmic reticulum-ATPase-2a). These results demonstrate that increased levels of miR-133a in the DM heart could prevent cardiac remodeling. Our P-V loop analysis showed a trend of decreased cardiac output, stroke volume, and ± dp/dt in Akita, which were blunted in Akita/miR-133aTg heart. These findings suggest that 13-15 week Akita heart undergoes adverse remodeling toward cardiomyopathy, which is prevented by miR-133a overexpression. In addition, increased cardiac miR-133a in the Akita heart did not change blood glucose levels but decreased lipid accumulation in the heart, suggesting inhibition of metabolic remodeling in the heart. Thus, miR-133a could be a promising therapeutic candidate to prevent DMCM.
Collapse
Affiliation(s)
- Tyler N Kambis
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid R Shahshahan
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Santosh K Yadav
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Yamashita Y, Oishi Y, Motomatsu Y, Hirayama K, Harada T, Ushijima T, Fujita S, Kimura S, Sonoda H, Tatewaki H, Tanoue Y, Sunagawa G, Nishikawa T, Saku K, Shiose A. Thoracic endografting increases cardiac afterload and leads to left ventricular hypertrophy in dogs. Eur J Cardiothorac Surg 2018; 55:618-625. [DOI: 10.1093/ejcts/ezy402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/19/2018] [Accepted: 10/13/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yoshiyuki Yamashita
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yasuhisa Oishi
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuma Motomatsu
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kazuto Hirayama
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takeaki Harada
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tomoki Ushijima
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Satoshi Fujita
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Satoshi Kimura
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hiromichi Sonoda
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hideki Tatewaki
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshihisa Tanoue
- Department of Advanced Cardiopulmonary Failure, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Genya Sunagawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takuya Nishikawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keita Saku
- Department of Advanced Risk Stratification for Cardiovascular Diseases, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka, Japan
| | - Akira Shiose
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
11
|
Boehme J, Le Moan N, Kameny RJ, Loucks A, Johengen MJ, Lesneski AL, Gong W, Goudy BD, Davis T, Tanaka K, Davis A, He Y, Long-Boyle J, Ivaturi V, Gobburu JVS, Winger JA, Cary SP, Datar SA, Fineman JR, Krtolica A, Maltepe E. Preservation of myocardial contractility during acute hypoxia with OMX-CV, a novel oxygen delivery biotherapeutic. PLoS Biol 2018; 16:e2005924. [PMID: 30335746 PMCID: PMC6193608 DOI: 10.1371/journal.pbio.2005924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 09/13/2018] [Indexed: 01/05/2023] Open
Abstract
The heart exhibits the highest basal oxygen (O2) consumption per tissue mass of any organ in the body and is uniquely dependent on aerobic metabolism to sustain contractile function. During acute hypoxic states, the body responds with a compensatory increase in cardiac output that further increases myocardial O2 demand, predisposing the heart to ischemic stress and myocardial dysfunction. Here, we test the utility of a novel engineered protein derived from the heme-based nitric oxide (NO)/oxygen (H-NOX) family of bacterial proteins as an O2 delivery biotherapeutic (Omniox-cardiovascular [OMX-CV]) for the hypoxic myocardium. Because of their unique binding characteristics, H-NOX–based variants effectively deliver O2 to hypoxic tissues, but not those at physiologic O2 tension. Additionally, H-NOX–based variants exhibit tunable binding that is specific for O2 with subphysiologic reactivity towards NO, circumventing a significant toxicity exhibited by hemoglobin (Hb)-based O2 carriers (HBOCs). Juvenile lambs were sedated, mechanically ventilated, and instrumented to measure cardiovascular parameters. Biventricular admittance catheters were inserted to perform pressure-volume (PV) analyses. Systemic hypoxia was induced by ventilation with 10% O2. Following 15 minutes of hypoxia, the lambs were treated with OMX-CV (200 mg/kg IV) or vehicle. Acute hypoxia induced significant increases in heart rate (HR), pulmonary blood flow (PBF), and pulmonary vascular resistance (PVR) (p < 0.05). At 1 hour, vehicle-treated lambs exhibited severe hypoxia and a significant decrease in biventricular contractile function. However, in OMX-CV–treated animals, myocardial oxygenation was improved without negatively impacting systemic or PVR, and both right ventricle (RV) and left ventricle (LV) contractile function were maintained at pre-hypoxic baseline levels. These data suggest that OMX-CV is a promising and safe O2 delivery biotherapeutic for the preservation of myocardial contractility in the setting of acute hypoxia. While hemoglobin is the primary oxygen delivery molecule used to maintain tissue oxygenation in metazoans, many organisms have other heme-containing proteins that can bind oxygen and other diatomic gases. Here, we tested whether a member of the H-NOX family of heme-containing proteins found in the thermostable bacterium Thermoanaerobacter tengcongensis can be engineered to deliver oxygen to severely hypoxic tissues in large mammals. This class of molecules has the advantage of high oxygen affinity and minimal nitric oxide reactivity. We demonstrate that these molecules can effectively deliver oxygen to a lamb heart with induced severe hypoxia, without overexposing the animal to oxygen or triggering systemic vascular reactivity. These molecules thus represent a novel class of oxygen delivery biotherapeutics to specifically target hypoxic tissue beds without the toxicity concerns of hemoglobin-based oxygen carriers. As tissue hypoxia is a central feature of many disease processes, this therapeutic approach may have broad clinical applicability.
Collapse
Affiliation(s)
- Jason Boehme
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Natacha Le Moan
- Omniox, Inc., San Carlos, California, United States of America
| | - Rebecca J. Kameny
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | | | - Michael J. Johengen
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Amy L. Lesneski
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Wenhui Gong
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Brian D. Goudy
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Tina Davis
- Omniox, Inc., San Carlos, California, United States of America
| | - Kevin Tanaka
- Omniox, Inc., San Carlos, California, United States of America
| | - Andrew Davis
- Omniox, Inc., San Carlos, California, United States of America
| | - Youping He
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Janel Long-Boyle
- Department of Clinical Pharmacology, University of California, San Francisco, San Francisco, California, United States of America
- Initiative for Pediatric Drug and Device Development (iPD3), San Francisco, California, United States of America
| | - Vijay Ivaturi
- Initiative for Pediatric Drug and Device Development (iPD3), San Francisco, California, United States of America
- School of Pharmacy, University of Maryland, Baltimore, United States of America
| | - Jogarao V. S. Gobburu
- Initiative for Pediatric Drug and Device Development (iPD3), San Francisco, California, United States of America
- School of Pharmacy, University of Maryland, Baltimore, United States of America
| | | | - Stephen P. Cary
- Omniox, Inc., San Carlos, California, United States of America
| | - Sanjeev A. Datar
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Initiative for Pediatric Drug and Device Development (iPD3), San Francisco, California, United States of America
| | - Ana Krtolica
- Omniox, Inc., San Carlos, California, United States of America
- * E-mail: (AK); (EM)
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Initiative for Pediatric Drug and Device Development (iPD3), San Francisco, California, United States of America
- * E-mail: (AK); (EM)
| |
Collapse
|
12
|
Prominent differences in left ventricular performance and myocardial properties between right ventricular and left ventricular-based pacing modes in rats. Sci Rep 2017; 7:5931. [PMID: 28725029 PMCID: PMC5517524 DOI: 10.1038/s41598-017-06197-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/30/2017] [Indexed: 01/16/2023] Open
Abstract
Biventricular pacing is an important modality to improve left ventricular (LV) synchronization and long-term function. However, the biological effects of this treatment are far from being elucidated and existing animal models are limited and demanding. Recently, we introduced an implanted device for double-site epicardial pacing in rats and echocardiographically demonstrated favorable effects of LV and biventricular (LV-based) pacing modes typically observed in humans. Here, this new animal model was further characterized. Electrodes were implanted either on the right atria (RA) and right ventricle (RV) or on the RV and LV. Following recovery, rats were either used for invasive hemodynamic measurements (pressure-volume analysis) or exposed to sustained RV vs. biventricular tachypacing for 3 days. RV pacing compromised, while LV-based pacing modes markedly enhanced cardiac performance. Changes in LV performance were associated with prominent compensatory changes in arterial resistance. Sustained RV tachypacing increased the electrocardiogram QTc interval by 7.9 ± 3.1 ms (n = 6, p < 0.05), dispersed refractoriness between the right and left pacing sites and induced important molecular changes mainly in the early-activated septal tissue. These effects were not observed during biventricular tachypacing (n = 6). Our results demonstrate that the rat is an attractive new model to study the biological consequences of LV dyssynchrony and resynchronization.
Collapse
|
13
|
Validation of a defibrillation lead ventricular volume measurement compared to three-dimensional echocardiography. Heart Rhythm 2017; 14:1515-1522. [PMID: 28603000 DOI: 10.1016/j.hrthm.2017.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND There is increasing evidence that using frequent invasive measures of pressure in patients with heart failure results in improved outcomes compared to traditional measures. Admittance, a measure of volume derived from preexisting defibrillation leads, is proposed as a new technique to monitor cardiac hemodynamics in patients with an implantable defibrillator. OBJECTIVE The purpose of this study was to evaluate the accuracy of a new ventricular volume sensor (VVS, CardioVol) compared with 3-dimenssional echocardiography (echo) in patients with an implantable defibrillator. METHODS Twenty-two patients referred for generator replacement had their defibrillation lead attached to VVS to determine the level of agreement to a volume measurement standard (echo). Two opposite hemodynamic challenges were sequentially applied to the heart (overdrive pacing and dobutamine administration) to determine whether real changes in hemodynamics could be reliably and repeatedly assessed with VVS. Equivalence of end-diastolic volume (EDV) and stroke volume (SV) determined by both methods was also assessed. RESULTS EDV and SV were compared using VVS and echo. VVS tracked expected physiologic trends. EDV was modulated -10% by overdrive pacing (14 mL). SV was modulated -13.7% during overdrive pacing (-6 mL) and increased over baseline +14.6% (+8 mL) with dobutamine. VVS and echo mean EDVs were found statistically equivalent, with margin of equivalence 13.8 mL (P <.05). Likewise, mean SVs were found statistically equivalent with margin of equivalence 15.8 mL (P <.05). CONCLUSION VVS provides an accurate method for ventricular volume assessment using chronically implanted defibrillator leads and is statistically equivalent to echo determination of mean EDV and SV.
Collapse
|
14
|
Silber D, Lachmann J. Invasive Hemodynamics of Pulmonary Disease and the Right Ventricle. Interv Cardiol Clin 2017; 6:329-343. [PMID: 28600088 DOI: 10.1016/j.iccl.2017.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Pulmonary hypertension (PH) falls into 5 groups, as defined by the World Health Organization. Swan-Ganz catheters determine precapillary versus postcapillary PH. The hemodynamic values of PH at rest and with vasodilatory challenge categorize the etiology of PH and guide treatment. RV maladaptations to increased pulmonary vascular resistance (PVR) and the chronicity of the right ventricle's (RV) response to increased PH and/or increased PVR can be understood with pressure-volume (PV) loops constructed with use of conductance catheters. These PV loops demonstrate the RV's ability to increase stroke volume in acutely and chronically increased PVR.
Collapse
Affiliation(s)
- David Silber
- Winthrop Cardiology Associates, PC, NYU-Winthrop Hospital, 212 Jericho Turnpike, Mineola, NY 11501, USA.
| | - Justine Lachmann
- Winthrop Cardiology Associates, PC, NYU-Winthrop Hospital, 212 Jericho Turnpike, Mineola, NY 11501, USA
| |
Collapse
|
15
|
Davidson S, Pretty C, Pironet A, Kamoi S, Balmer J, Desaive T, Chase JG. Minimally invasive, patient specific, beat-by-beat estimation of left ventricular time varying elastance. Biomed Eng Online 2017; 16:42. [PMID: 28407773 PMCID: PMC5390429 DOI: 10.1186/s12938-017-0338-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/06/2017] [Indexed: 01/13/2023] Open
Abstract
Background The aim of this paper was to establish a minimally invasive method for deriving the left ventricular time varying elastance (TVE) curve beat-by-beat, the monitoring of which’s inter-beat evolution could add significant new data and insight to improve diagnosis and treatment. The method developed uses the clinically available inputs of aortic pressure, heart rate and baseline end-systolic volume (via echocardiography) to determine the outputs of left ventricular pressure, volume and dead space volume, and thus the TVE curve. This approach avoids directly assuming the shape of the TVE curve, allowing more effective capture of intra- and inter-patient variability. Results The resulting TVE curve was experimentally validated against the TVE curve as derived from experimentally measured left ventricular pressure and volume in animal models, a data set encompassing 46,318 heartbeats across 5 Piétrain pigs. This simulated TVE curve was able to effectively approximate the measured TVE curve, with an overall median absolute error of 11.4% and overall median signed error of −2.5%. Conclusions The use of clinically available inputs means there is potential for real-time implementation of the method at the patient bedside. Thus the method could be used to provide additional, patient specific information on intra- and inter-beat variation in heart function. Electronic supplementary material The online version of this article (doi:10.1186/s12938-017-0338-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shaun Davidson
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand.
| | - Chris Pretty
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Antoine Pironet
- GIGA-Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Shun Kamoi
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Joel Balmer
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Thomas Desaive
- GIGA-Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - J Geoffrey Chase
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
16
|
Promising approach for the preclinical assessment of cardiac risks using left ventricular pressure-volume loop analyses in anesthetized monkeys. J Pharmacol Toxicol Methods 2017; 84:1-10. [DOI: 10.1016/j.vascn.2016.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 11/23/2022]
|
17
|
Golob MJ, Wang Z, Prostrollo AJ, Hacker TA, Chesler NC. Limiting collagen turnover via collagenase-resistance attenuates right ventricular dysfunction and fibrosis in pulmonary arterial hypertension. Physiol Rep 2016; 4:4/11/e12815. [PMID: 27252252 PMCID: PMC4908492 DOI: 10.14814/phy2.12815] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 12/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe form of pulmonary hypertension in which right ventricular (RV) afterload is increased and death typically occurs due to decompensated RV hypertrophy and failure. Collagen accumulation has been implicated in pulmonary artery remodeling, but how it affects RV performance remains unclear. Here, we sought to identify the role of collagen turnover, defined as the balance between collagen synthesis and degradation, in RV structure and function in PAH. To do so, we exposed mutant (Col1a1R/R) mice, in which collagen type I degradation is impaired such that collagen turnover is reduced, and wild‐type (Col1a1+/+) littermates to 14 days of chronic hypoxia combined with SUGEN treatment (HySu) to recapitulate characteristics of clinical PAH. RV structure and function were measured by echocardiography, RV catheterization, and histology. Despite comparable increases in RV systolic pressure (Col1a1+/+: 46 ± 2 mmHg; Col1a1R/R: 47 ± 3 mmHg), the impaired collagen degradation in Col1a1R/R mice resulted in no RV collagen accumulation, limited RV hypertrophy, and maintained right ventricular‐pulmonary vascular coupling with HySu exposure. The preservation of cardiac function in the mutant mice indicates a beneficial role of limited collagen turnover via impaired degradation in RV remodeling in response to chronic pressure overload. Our results suggest novel treatments that reduce collagen turnover may offer a new therapeutic strategy for PAH patients.
Collapse
Affiliation(s)
- Mark J Golob
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, Wisconsin Materials Science Program, University of Wisconsin-Madison College of Engineering, Madison, Wisconsin
| | - Zhijie Wang
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, Wisconsin
| | - Anthony J Prostrollo
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, Wisconsin
| | - Timothy A Hacker
- Department of Medicine, Medical Science Center University of Wisconsin-Madison, Madison, Wisconsin
| | - Naomi C Chesler
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, Wisconsin
| |
Collapse
|
18
|
Axelsson B, Häggmark S, Svenmarker S, Johansson G, Gupta A, Tydén H, Wouters P, Haney M. Effects of Combined Milrinone and Levosimendan Treatment on Systolic and Diastolic Function During Postischemic Myocardial Dysfunction in a Porcine Model. J Cardiovasc Pharmacol Ther 2016; 21:495-503. [DOI: 10.1177/1074248416628675] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/28/2015] [Indexed: 11/17/2022]
Abstract
It is not known whether there are positive or negative interactions on ventricular function when a calcium-sensitizing inotrope is added to a phosphodiesterase inhibitor in the clinical setting of acute left ventricular (LV) dysfunction. We hypothesized that when levosimendan is added to milrinone treatment, there will be synergetic inotropic and lusitropic effects. This was tested in an anesthetized porcine postischemic global LV injury model, where ventricular pressures and volumes (conductance volumetry) were measured. A global ischemic injury was induced by repetitive left main stem coronary artery occlusions. Load-independent indices of LV function were assessed before and after ventricular injury, after milrinone treatment, and finally after addition of levosimendan to the milrinone treatment. Nonparametric, within-group comparisons were made. The protocol was completed in 12 pigs, 7 of which received the inotrope treatment and 5 of which served as controls. Milrinone led to positive lusitropic effects seen by improvement in tau after myocardial stunning. The addition of levosimendan to milrinone further increased lusitropic state. The latter effect could however not be attributed solely to levosimendan, since lusitropic state also improved spontaneously in time-matched controls at the same rate during the corresponding period. When levosimendan was added to milrinone infusion, there was no increase in systolic function (preload recruitable stroke work) compared to milrinone treatment alone. We conclude that in this model of postischemic LV dysfunction, there appears to be no clear improvement in systolic or diastolic function after addition of levosimendan to established milrinone treatment but also no negative effects of levosimendan in this context.
Collapse
Affiliation(s)
- Birger Axelsson
- Department of Cardiovascular and Thoracic Surgery, Örebro University Hospital, Örebro, Sweden
- Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Sören Häggmark
- Department of Surgical and Perioperative Sciences, Heart Centre and Anesthesiology and Intensive Care Medicine, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Staffan Svenmarker
- Department of Surgical and Perioperative Sciences, Heart Centre and Anesthesiology and Intensive Care Medicine, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Göran Johansson
- Department of Surgical and Perioperative Sciences, Anesthesiology and Intensive Care Medicine, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Anil Gupta
- Department of Anesthesiology and Intensive Care, and Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Hans Tydén
- Department of Cardiovascular and Thoracic Surgery, Örebro University Hospital, Örebro and Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Patrick Wouters
- Department of Anesthesiology, University Hospital Ghent, Ghent, Belgium
| | - Michael Haney
- Department of Surgical and Perioperative Sciences, Anesthesiology and Intensive Care Medicine, Faculty of Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
19
|
Effects of adrenaline on circulatory dynamics and cardiac function in rats administered chlorpromazine. Odontology 2016; 105:103-107. [PMID: 27040284 DOI: 10.1007/s10266-016-0241-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 03/07/2016] [Indexed: 10/22/2022]
Abstract
We aimed to elucidate changes in circulatory dynamics and cardiac function during concomitant use of chlorpromazine (CPZ) and adrenaline (AD). An arterial line and left intraventricular pressure-volume measurement catheter were inserted in rats. CPZ 10 mg/kg was administered to the left great adductor muscle, followed by normal saline (NS) or AD 50 μg/kg through the tongue 20 min later. End-diastolic volume (V ed), end-systolic pressure (P es), stroke volume (SV), stroke work (SW), end-systolic volume elastance (E es), systolic blood pressure (SBP), diastolic blood pressure (DBP), and pulse rate (PR) were measured. Following AD administration, V ed significantly decreased at 2-4 and 10 min than that in control rats; P es significantly decreased at 1 min; E es significantly increased from 2 to 10 min; SV did not change significantly, and SW significantly reduced at 1 and 2 min; SBP and DBP were lower at 1-3 min than in the control; and PR increased at 10 min. These findings suggest that when AD-containing local anesthetics are administered during dental treatment of patients taking CPZ, there is a risk of a temporary drop in blood pressure. However, the blood pressure is recovered a few minutes later by the increase in afterload and the myocardial contractile force.
Collapse
|
20
|
Liu R, Correll RN, Davis J, Vagnozzi RJ, York AJ, Sargent MA, Nairn AC, Molkentin JD. Cardiac-specific deletion of protein phosphatase 1β promotes increased myofilament protein phosphorylation and contractile alterations. J Mol Cell Cardiol 2015; 87:204-13. [PMID: 26334248 PMCID: PMC4637224 DOI: 10.1016/j.yjmcc.2015.08.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 12/27/2022]
Abstract
There are 3 protein phosphatase 1 (PP1) catalytic isoforms (α, β and γ) encoded within the mammalian genome. These 3 gene products share ~90% amino acid homology within their catalytic domains but each has unique N- and C-termini that likely underlie distinctive subcellular localization or functionality. In this study, we assessed the effect associated with the loss of each PP1 isoform in the heart using a conditional Cre-loxP targeting approach in mice. Ppp1ca-loxP, Ppp1cb-loxP and Ppp1cc-loxP alleles were crossed with either an Nkx2.5-Cre knock-in containing allele for early embryonic deletion or a tamoxifen inducible α-myosin heavy chain (αMHC)-MerCreMer transgene for adult and cardiac-specific deletion. We determined that while deletion of Ppp1ca (PP1α) or Ppp1cc (PP1γ) had little effect on the whole heart, deletion of Ppp1cb (PP1β) resulted in concentric remodeling of the heart, interstitial fibrosis and contractile dysregulation, using either the embryonic or adult-specific Cre-expressing alleles. However, myocytes isolated from Ppp1cb deleted hearts surprisingly showed enhanced contractility. Mechanistically we found that deletion of any of the 3 PP1 gene-encoding isoforms had no effect on phosphorylation of phospholamban, nor were Ca(2+) handling dynamics altered in adult myocytes from Ppp1cb deleted hearts. However, the loss of Ppp1cb from the heart, but not Ppp1ca or Ppp1cc, resulted in elevated phosphorylation of myofilament proteins such as myosin light chain 2 and cardiac myosin binding protein C, consistent with an enriched localization profile of this isoform to the sarcomeres. These results suggest a unique functional role for the PP1β isoform in affecting cardiac contractile function.
Collapse
Affiliation(s)
- Ruijie Liu
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Robert N Correll
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Jennifer Davis
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Ronald J Vagnozzi
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Allen J York
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, OH 45229, USA.
| |
Collapse
|
21
|
Abraham D, Mao L. Cardiac Pressure-Volume Loop Analysis Using Conductance Catheters in Mice. J Vis Exp 2015. [PMID: 26436838 DOI: 10.3791/52942] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac pressure-volume loop analysis is the "gold-standard" in the assessment of load-dependent and load-independent measures of ventricular systolic and diastolic function. Measures of ventricular contractility and compliance are obtained through examination of cardiac response to changes in afterload and preload. These techniques were originally developed nearly three decades ago to measure cardiac function in large mammals and humans. The application of these analyses to small mammals, such as mice, has been accomplished through the optimization of microsurgical techniques and creation of conductance catheters. Conductance catheters allow for estimation of the blood pool by exploiting the relationship between electrical conductance and volume. When properly performed, these techniques allow for testing of cardiac function in genetic mutant mouse models or in drug treatment studies. The accuracy and precision of these studies are dependent on careful attention to the calibration of instruments, systematic conduct of hemodynamic measurements and data analyses. We will review the methods of conducting pressure-volume loop experiments using a conductance catheter in mice.
Collapse
Affiliation(s)
| | - Lan Mao
- Department of Medicine, Duke University Medical Center
| |
Collapse
|
22
|
Chaanine AH, Gordon RE, Nonnenmacher M, Kohlbrenner E, Benard L, Hajjar RJ. High-dose chloroquine is metabolically cardiotoxic by inducing lysosomes and mitochondria dysfunction in a rat model of pressure overload hypertrophy. Physiol Rep 2015; 3:3/7/e12413. [PMID: 26152691 PMCID: PMC4552516 DOI: 10.14814/phy2.12413] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Autophagy, macroautophagy and chaperone-mediated autophagy (CMA), are upregulated in pressure overload (PO) hypertrophy. In this study, we targeted this process at its induction using 3 methyladenine and at the lysosomal level using chloroquine and evaluated the effects of these modulations on cardiac function and myocyte ultrastructure. Sprague–Dawley rats weighing 200 g were subjected to ascending aortic banding. After 1 week of PO, animals were randomized to receive 3 methyladenine versus chloroquine, intraperitoneally, for 2 weeks at a dose of 40 and 50 mg/kg/day, respectively. Saline injection was used as control. Chloroquine treatment, in PO, resulted in regression in cardiac hypertrophy but with significant impairments in cardiac relaxation and contractility. Ultrastructurally, chloroquine accentuated mitochondrial fragmentation and cristae destruction with a plethora of autophagosomes containing collapsed mitochondria and lysosomal lamellar bodies. In contrast, 3 methyladenine improved cardiac function and attenuated mitochondrial fragmentation and autophagososme formation. Markers of macroautophagy and CMA were significantly decreased in the chloroquine group; whereas 3 methyladenine treatment significantly attenuated macroautophagy with a compensatory increase in CMA. Furthermore, chloroquine accentuated PO induced oxidative stress through the further decrease in the expression of manganese superoxide dismutase; whereas, 3 MA had a completely opposite effect. Taken together, these data suggest that high-dose chloroquine, in addition to its effect on the autophagy-lysosome pathway, significantly impairs mitochondrial antioxidant buffering capacity and accentuates oxidative stress and mitochondrial dysfunction in PO hypertrophy; highlighting, the cautious administration of this drug in high oxidative stress conditions, such as pathological hypertrophy or heart failure.
Collapse
Affiliation(s)
- Antoine H Chaanine
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York
| | - Ronald E Gordon
- Pathology Department, Mount Sinai School of Medicine, New York, New York
| | | | - Erik Kohlbrenner
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York
| | - Ludovic Benard
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York
| | - Roger J Hajjar
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
23
|
Jonker SS, Giraud GD, Espinoza HM, Davis EN, Crossley DA. Effects of chronic hypoxia on cardiac function measured by pressure-volume catheter in fetal chickens. Am J Physiol Regul Integr Comp Physiol 2015; 308:R680-9. [PMID: 25652537 DOI: 10.1152/ajpregu.00484.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/02/2015] [Indexed: 11/22/2022]
Abstract
Hypoxia is a common component of many developmental insults and has been studied in early-stage chicken development. However, its impact on cardiac function and arterial-ventricular coupling in late-stage chickens is relatively unknown. To test the hypothesis that hypoxic incubation would reduce baseline cardiac function but protect the heart during acute hypoxia in late-stage chickens, white Leghorn eggs were incubated at 21% O2 or 15% O2. At 90% of incubation (19 days), hypoxic incubation caused growth restriction (-20%) and increased the LV-to-body ratio (+41%). Left ventricular (LV) pressure-volume loops were measured in anesthetized chickens in normoxia and acute hypoxia (10% O2). Hypoxic incubation lowered the maximal rate of pressure generation (ΔP/ΔtMax; -22%) and output (-57%), whereas increasing end-systolic elastance (ELV; +31%) and arterial elastance (EA; +122%) at similar heart rates to normoxic incubation. Both hypoxic incubation and acute hypoxia lengthened the half-time of relaxation (τ; +24%). Acute hypoxia reduced heart rate (-8%) and increased end-diastolic pressure (+35%). Hearts were collected for mRNA analysis. Hypoxic incubation was marked by decreased mRNA expression of sarco(endo)plasmic reticulum Ca(2+)-ATPase 2, Na(+)/Ca(2+) exchanger 1, phospholamban, and ryanodine receptor. In summary, hypoxic incubation reduces LV function in the late-stage chicken by slowing pressure generation and relaxation, which may be driven by altered intracellular excitation-contraction coupling. Cardiac efficiency is greatly reduced after hypoxic incubation. In both incubation groups acute hypoxia reduced diastolic function.
Collapse
Affiliation(s)
- Sonnet S Jonker
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon;
| | - George D Giraud
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon; Veterans Affairs Portland Health Care System, Portland, Oregon; and
| | - Herbert M Espinoza
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Erica N Davis
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Dane A Crossley
- Department of Biological Sciences, University of North Texas, Denton, Texas
| |
Collapse
|
24
|
Granfeldt A, Letson HL, Dobson GP, Shi W, Vinten-Johansen J, Tønnesen E. Adenosine, lidocaine and Mg2+ improves cardiac and pulmonary function, induces reversible hypotension and exerts anti-inflammatory effects in an endotoxemic porcine model. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:682. [PMID: 25497775 PMCID: PMC4301798 DOI: 10.1186/s13054-014-0682-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/20/2014] [Indexed: 12/24/2022]
Abstract
Introduction The combination of Adenosine (A), lidocaine (L) and Mg2+ (M) (ALM) has demonstrated cardioprotective and resuscitative properties in models of cardiac arrest and hemorrhagic shock. This study evaluates whether ALM also demonstrates organ protective properties in an endotoxemic porcine model. Methods Pigs (37 to 42 kg) were randomized into: 1) Control (n = 8) or 2) ALM (n = 8) followed by lipopolysaccharide infusion (1 μg∙kg-1∙h-1) for five hours. ALM treatment consisted of 1) a high dose bolus (A (0.82 mg/kg), L (1.76 mg/kg), M (0.92 mg/kg)), 2) one hour continuous infusion (A (300 μg∙kg-1 ∙min-1), L (600 μg∙kg-1 ∙min-1), M (336 μg∙kg-1 ∙min-1)) and three hours at a lower dose (A (240∙kg-1∙min-1), L (480 μg∙kg-1∙min-1), M (268 μg∙kg-1 ∙min-1)); controls received normal saline. Hemodynamic, cardiac, pulmonary, metabolic and renal functions were evaluated. Results ALM lowered mean arterial pressure (Mean value during infusion period: ALM: 47 (95% confidence interval (CI): 44 to 50) mmHg versus control: 79 (95% CI: 75 to 85) mmHg, P <0.0001). After cessation of ALM, mean arterial pressure immediately increased (end of study: ALM: 88 (95% CI: 81 to 96) mmHg versus control: 86 (95% CI: 79 to 94) mmHg, P = 0.72). Whole body oxygen consumption was significantly reduced during ALM infusion (ALM: 205 (95% CI: 192 to 217) ml oxygen/min versus control: 231 (95% CI: 219 to 243) ml oxygen/min, P = 0.016). ALM treatment reduced pulmonary injury evaluated by PaO2/FiO2 ratio (ALM: 388 (95% CI: 349 to 427) versus control: 260 (95% CI: 221 to 299), P = 0.0005). ALM infusion led to an increase in heart rate while preserving preload recruitable stroke work. Creatinine clearance was significantly lower during ALM infusion but reversed after cessation of infusion. ALM reduced tumor necrosis factor-α peak levels (ALM 7121 (95% CI: 5069 to 10004) pg/ml versus control 11596 (95% CI: 9083 to 14805) pg/ml, P = 0.02). Conclusion ALM infusion induces a reversible hypotensive and hypometabolic state, attenuates tumor necrosis factor-α levels and improves cardiac and pulmonary function, and led to a transient drop in renal function that was reversed after the treatment was stopped.
Collapse
Affiliation(s)
- Asger Granfeldt
- Department of Anesthesiology, Aarhus University Hospital, Nørrebrogade 44 building 21 1st floor 8000, Aarhus, Denmark. .,Department of Anesthesiology, Regional Hospital of Randers, Skovlyvej 1, 8930, Randers, Denmark.
| | - Hayley L Letson
- Heart, Trauma & Sepsis Research Laboratory, Australian Institute of Tropical Health and Medicine, School of Medicine and Dentistry, James Cook University, Pharmacy and Medical Research Building 47, Rm 113B, Townsville, Queensland, Australia.
| | - Geoffrey P Dobson
- Heart, Trauma & Sepsis Research Laboratory, Australian Institute of Tropical Health and Medicine, School of Medicine and Dentistry, James Cook University, Pharmacy and Medical Research Building 47, Rm 113B, Townsville, Queensland, Australia.
| | - Wei Shi
- The Cardiothoracic Research Laboratory, Carlyle Fraser Heart Center, Emory University School of Medicine, 387 Technology Circle Suite 180, Atlanta, Georgia 30313, USA.
| | - Jakob Vinten-Johansen
- The Cardiothoracic Research Laboratory, Carlyle Fraser Heart Center, Emory University School of Medicine, 387 Technology Circle Suite 180, Atlanta, Georgia 30313, USA.
| | - Else Tønnesen
- Department of Anesthesiology, Aarhus University Hospital, Nørrebrogade 44 building 21 1st floor 8000, Aarhus, Denmark.
| |
Collapse
|
25
|
Schreier DA, Hacker TA, Hunter K, Eickoff J, Liu A, Song G, Chesler N. Impact of increased hematocrit on right ventricular afterload in response to chronic hypoxia. J Appl Physiol (1985) 2014; 117:833-9. [PMID: 25170068 DOI: 10.1152/japplphysiol.00059.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic hypoxia causes chronic mountain sickness through hypoxia-induced pulmonary hypertension (HPH) and increased hematocrit. Here, we investigated the impact of increased hematocrit and HPH on right ventricular (RV) afterload via pulmonary vascular impedance. Mice were exposed to chronic normobaric hypoxia (10% oxygen) for 10 (10H) or 21 days (21H). After baseline hemodynamic measurements, ∼500 μl of blood were extracted and replaced with an equal volume of hydroxyethylstarch to normalize hematocrit and all hemodynamic measurements were repeated. In addition, ∼500 μl of blood were extracted and replaced in control mice with an equal volume of 90% hematocrit blood. Chronic hypoxia increased input resistance (Z0 increased 82% in 10H and 138% in 21H vs. CTL; P < 0.05) and characteristic impedance (ZC increased 76% in 10H and 109% in 21H vs. CTL; P < 0.05). Hematocrit normalization did not decrease mean pulmonary artery pressure but did increase cardiac output such that both Z0 and ZC decreased toward control levels. Increased hematocrit in control mice did not increase pressure but did decrease cardiac output such that Z0 increased. The paradoxical decrease in ZC with an acute drop in hematocrit and no change in pressure are likely due to inertial effects secondary to the increase in cardiac output. A novel finding of this study is that an increase in hematocrit affects the pulsatile RV afterload in addition to the steady RV afterload (Z0). Furthermore, our results highlight that the conventional interpretation of ZC as a measure of proximal artery stiffness is not valid in all physiological and pathological states.
Collapse
Affiliation(s)
- David A Schreier
- Department of Biomedical Engineering University of Wisconsin, Madison, Wisconson
| | - Timothy A Hacker
- Department of Medicine Medical Science Center, Madison, Wisconsin; and
| | - Kendall Hunter
- Department of Bioengineering University of Colorado, Aurora, Colorado
| | - Jens Eickoff
- Department of Medicine Medical Science Center, Madison, Wisconsin; and
| | - Aiping Liu
- Department of Biomedical Engineering University of Wisconsin, Madison, Wisconson
| | - Gouqing Song
- Department of Medicine Medical Science Center, Madison, Wisconsin; and
| | - Naomi Chesler
- Department of Biomedical Engineering University of Wisconsin, Madison, Wisconson; Department of Medicine Medical Science Center, Madison, Wisconsin; and
| |
Collapse
|
26
|
Larson ER, Porterfield JE, Sagar S, Marmol-Velez J, Panday M, Escobedo D, Michalek J, Ouyang Y, Valvano JW, Pearce JA, Feldman MD. Admittance to detect alterations in left ventricular stroke volume. Heart Rhythm 2014; 11:2075-83. [PMID: 24981870 DOI: 10.1016/j.hrthm.2014.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Implantable cardioverter-defibrillators monitor intracardiac electrograms (EGMs) to discriminate between ventricular and supraventricular tachycardias. The incidence of inappropriate shocks remains high because of misclassification of the tachycardia in an otherwise hemodynamically stable individual. Coupling EGMs with an assessment of left ventricular (LV) stroke volume (SV) could help in gauging hemodynamics during an arrhythmia and reducing inappropriate shocks. OBJECTIVE The purpose of this study was to use the admittance method to accurately derive LV SV. METHODS Ultrasonic flow probe and LV endocardial crystals were used in canines (n = 12) as the standard for LV SV. Biventricular pacing leads were inserted to obtain admittance measurements. A tetrapolar, complex impedance measurement was made between the Bi-V leads. The real and imaginary components of impedance were used to discard the myocardial component from the blood component to derive instantaneous blood conductance (Gb). Alterations in SV were measured during right ventricular pacing, dopamine infusion, and inferior vena cava occlusion. RESULTS Gb tracks steady-state changes in SV more accurately than traditional magnitude (ie, |Y|, without removal of the muscle signal) during right ventricular pacing and dopamine infusion (P = .004). Instantaneous LV volume also was tracked more accurately by Gb than ∣Y∣ in the subset of subjects that underwent inferior vena cava occlusions (n = 5, P = .025). Finite element modeling demonstrates that admittance shifts more sensitivity of the measurement to the LV blood chamber as the mechanism for improvement (see Online Appendix). CONCLUSION Monitoring LV SV is possible using the admittance method with biventricular pacing leads. The technique could be piggybacked to complement EGMs to determine if arrhythmias are hemodynamically unstable.
Collapse
Affiliation(s)
- Erik R Larson
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas
| | | | - Sandeep Sagar
- Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Juan Marmol-Velez
- Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Manoj Panday
- Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Daniel Escobedo
- Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Joel Michalek
- Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Yongjian Ouyang
- Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jonathan W Valvano
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas
| | - John A Pearce
- Department of Electrical Engineering, University of Texas at Austin, Austin, Texas
| | - Marc D Feldman
- Admittance Technologies, Austin, Texas; Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas.
| |
Collapse
|
27
|
van Hout GPJ, Jansen of Lorkeers SJ, Gho JMIH, Doevendans PA, van Solinge WW, Pasterkamp G, Chamuleau SAJ, Hoefer IE. Admittance-based pressure-volume loops versus gold standard cardiac magnetic resonance imaging in a porcine model of myocardial infarction. Physiol Rep 2014; 2:e00287. [PMID: 24771693 PMCID: PMC4001878 DOI: 10.14814/phy2.287] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A novel admittance‐based pressure–volume system (AS) has recently been developed and introduced. Thus far, the new technique has been validated predominantly in small animals. In large animals it has only been compared to three‐dimensional echocardiography (3DE) where the AS showed to overestimate left ventricular (LV) volumes. To fully determine the accuracy of this device, we compared the AS with gold standard cardiac magnetic resonance imaging (CMRI) in a porcine model of chronic myocardial infarction (MI). Fourteen pigs were subjected to 90 min closed chest balloon occlusion of the left anterior descending artery. After 8 weeks of follow up, pigs were consecutively subjected to LV volume measurements by the AS, CMRI, and 3DE under general anesthesia. The AS overestimated end diastolic volume (EDV; +20.9 ± 30.6 mL, P = 0.024) and end systolic volume (ESV; +17.7 ± 29.4 mL, P = 0.042) but not ejection fraction (EF; +2.46 ± 6.16%, P = NS) compared to CMRI. Good correlations of EDV (R = 0.626, P = 0.017) and EF (R = 0.704, P = 0.005) between the AS and CMRI were observed. EF measured by the AS and 3DE also correlated significantly (R = 0.624, P = 0.030). After subjection of pigs to MI, the AS very moderately overestimates LV volumes and shows accurate measurements for EF compared to CMRI. This makes the AS a useful tool to determine cardiac function and dynamic changes in large animal models of cardiac disease. Is the novel admittance‐based pressure–volume loop system reliable for the assessment of left ventricular volumes compared to gold standard cardiac magnetic resonance imaging in a porcine model of myocardial infarction? In the postinfarction remodeled heart, admittance‐based pressure–volume loop measurements accurately measure ejection fraction and very moderately overestimate end diastolic and end systolic volumes compared to gold standard cardiac magnetic resonance imaging, making it a very useful technique for cardiac function assessment in experimental studies.
Collapse
Affiliation(s)
- Gerardus P J van Hout
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cook JR, Carta L, Bénard L, Chemaly ER, Chiu E, Rao SK, Hampton TG, Yurchenco P, Costa KD, Hajjar RJ, Ramirez F. Abnormal muscle mechanosignaling triggers cardiomyopathy in mice with Marfan syndrome. J Clin Invest 2014; 124:1329-39. [PMID: 24531548 PMCID: PMC3934180 DOI: 10.1172/jci71059] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 12/12/2013] [Indexed: 12/11/2022] Open
Abstract
Patients with Marfan syndrome (MFS), a multisystem disorder caused by mutations in the gene encoding the extracellular matrix (ECM) protein fibrillin 1, are unusually vulnerable to stress-induced cardiac dysfunction. The prevailing view is that MFS-associated cardiac dysfunction is the result of aortic and/or valvular disease. Here, we determined that dilated cardiomyopathy (DCM) in fibrillin 1-deficient mice is a primary manifestation resulting from ECM-induced abnormal mechanosignaling by cardiomyocytes. MFS mice displayed spontaneous emergence of an enlarged and dysfunctional heart, altered physical properties of myocardial tissue, and biochemical evidence of chronic mechanical stress, including increased angiotensin II type I receptor (AT1R) signaling and abated focal adhesion kinase (FAK) activity. Partial fibrillin 1 gene inactivation in cardiomyocytes was sufficient to precipitate DCM in otherwise phenotypically normal mice. Consistent with abnormal mechanosignaling, normal cardiac size and function were restored in MFS mice treated with an AT1R antagonist and in MFS mice lacking AT1R or β-arrestin 2, but not in MFS mice treated with an angiotensin-converting enzyme inhibitor or lacking angiotensinogen. Conversely, DCM associated with abnormal AT1R and FAK signaling was the sole abnormality in mice that were haploinsufficient for both fibrillin 1 and β1 integrin. Collectively, these findings implicate fibrillin 1 in the physiological adaptation of cardiac muscle to elevated workload.
Collapse
MESH Headings
- Adult
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/physiopathology
- Child
- Cross-Sectional Studies
- Extracellular Matrix/metabolism
- Fibrillin-1
- Fibrillins
- Focal Adhesion Kinase 1/metabolism
- Humans
- Losartan/pharmacology
- MAP Kinase Signaling System
- Male
- Marfan Syndrome/complications
- Marfan Syndrome/metabolism
- Marfan Syndrome/pathology
- Marfan Syndrome/physiopathology
- Mechanotransduction, Cellular
- Mice
- Mice, Transgenic
- Microfilament Proteins/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Organ Size
- Receptor, Angiotensin, Type 1/metabolism
Collapse
Affiliation(s)
- Jason R. Cook
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Luca Carta
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Ludovic Bénard
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Elie R. Chemaly
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Emily Chiu
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Satish K. Rao
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Thomas G. Hampton
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Peter Yurchenco
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | | | - Kevin D. Costa
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Roger J. Hajjar
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| | - Francesco Ramirez
- Department of Pharmacology and Systems Therapeutics and
Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, Massachusetts, USA.
Department of Pathology and Laboratory Medicine, Robert W. Johnson School of Medicine, Piscataway, New Jersey, USA
| |
Collapse
|
29
|
Chaanine AH, Nonnenmacher M, Kohlbrenner E, Jin D, Kovacic JC, Akar FG, Hajjar RJ, Weber T. Effect of bortezomib on the efficacy of AAV9.SERCA2a treatment to preserve cardiac function in a rat pressure-overload model of heart failure. Gene Ther 2014; 21:379-386. [PMID: 24572786 PMCID: PMC3976435 DOI: 10.1038/gt.2014.7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/24/2013] [Accepted: 01/09/2014] [Indexed: 01/14/2023]
Abstract
Adeno-associated virus (AAV)-based vectors are promising vehicles for therapeutic gene delivery, including for the treatment for heart failure. It has been demonstrated for each of the AAV serotypes 1 through 8 that inhibition of the proteasome results in increased transduction efficiencies. For AAV9, however, the effect of proteasome inhibitors on in vivo transduction has until now not been evaluated. Here we demonstrate, in a well-established rodent heart failure model, that concurrent treatment with the proteasome inhibitor bortezomib does not enhance the efficacy of AAV9.SERCA2a to improve cardiac function as examined by echocardiography and pressure volume analysis. Western blot analysis of SERCA2a protein and reverse transcription-PCR of SERCA2a mRNA demonstrated that bortezomib had no effect on either endogenous rat SERCA2a levels nor on expression levels of human SERCA2a delivered by AAV9.SERCA2a. Similarly, the number of AAV9 genomes in heart samples was unaffected by bortezomib treatment. Interestingly, whereas transduction of HeLa cells and neonatal rat cardiomyocytes by AAV9 was stimulated by bortezomib, transduction of adult rat cardiomyocytes was inhibited. These results indicate an organ/cell-type-specific effect of proteasome inhibition on AAV9 transduction. A future detailed analysis of the underlying molecular mechanisms promises to facilitate the development of improved AAV vectors.
Collapse
Affiliation(s)
- Antoine H Chaanine
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Mathieu Nonnenmacher
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dongzhu Jin
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jason C Kovacic
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Fadi G Akar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Thomas Weber
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
30
|
Wang Z, Schreier DA, Hacker TA, Chesler NC. Progressive right ventricular functional and structural changes in a mouse model of pulmonary arterial hypertension. Physiol Rep 2013; 1:e00184. [PMID: 24744862 PMCID: PMC3970737 DOI: 10.1002/phy2.184] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/13/2013] [Accepted: 10/18/2013] [Indexed: 11/18/2022] Open
Abstract
Right ventricle (RV) dysfunction occurs with progression of pulmonary arterial hypertension (PAH) due to persistently elevated ventricular afterload. A critical knowledge gap is the molecular mechanisms that govern the transition from RV adaptation to RV maladaptation, which leads to failure. Here, we hypothesize that the recently established mouse model of PAH, via hypoxia and SU5416 treatment (HySu), captures that transition from adaptive to maladaptive RV remodeling including impairments in RV function and decreases in the efficiency of RV interactions with the pulmonary vasculature. To test this hypothesis, we exposed C57BL6 male mice to 0 (control), 14, 21, and 28 days of HySu and then obtained synchronized RV pressure and volume measurements in vivo. With increasing HySu exposure duration, arterial afterload increased monotonically, leading to a continuous increase in RV stroke work, RV fibrosis, and RV wall stiffening (P < 0.05). RV contractility increased at 14 days of HySu exposure and then plateaued (P < 0.05). As a result, ventricular–vascular coupling efficiency tended to increase at 14 days and then decrease. Our results suggest that RV remodeling may begin to shift from adaptive to maladaptive with increasing duration of HySu exposure, which would mimic changes in RV function with PAH progression found clinically. However, for the duration of HySu exposure used here, no drop in cardiac output was found. We conclude that the establishment of a mouse model for overt RV failure due to PAH remains an important task. This article describes the progressive changes in mouse right ventricle (RV) structure and function during the pulmonary arterial hypertension development. The findings may shed light on the transition from adaptive to maladaptive RV remodeling, which eventually leads to failure.
Collapse
Affiliation(s)
- Zhijie Wang
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, 53706, Wisconsin
| | - David A Schreier
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, 53706, Wisconsin
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin, Madison, 53706, Wisconsin
| | - Naomi C Chesler
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, 53706, Wisconsin ; Department of Medicine, University of Wisconsin, Madison, 53706, Wisconsin
| |
Collapse
|
31
|
van Hout GPJ, de Jong R, Vrijenhoek JEP, Timmers L, Duckers HJ, Hoefer IE. Admittance-based pressure-volume loop measurements in a porcine model of chronic myocardial infarction. Exp Physiol 2013; 98:1565-75. [DOI: 10.1113/expphysiol.2013.074179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Schreier D, Hacker T, Song G, Chesler N. The role of collagen synthesis in ventricular and vascular adaptation to hypoxic pulmonary hypertension. J Biomech Eng 2013; 135:021018. [PMID: 23445063 DOI: 10.1115/1.4023480] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a rapidly fatal disease in which mortality is typically due to right ventricular (RV) failure. An excellent predictor of mortality in PAH is proximal pulmonary artery stiffening, which is mediated by collagen accumulation in hypoxia-induced pulmonary hypertension (HPH) in mice. We sought to investigate the impact of limiting vascular and ventricular collagen accumulation on RV function and the hemodynamic coupling efficiency between the RV and pulmonary vasculature. Inbred mice were exposed to chronic hypoxia for 10 days with either no treatment (HPH) or with treatment with a proline analog that impairs collagen synthesis (CHOP-PEG; HPH + CP). Both groups were compared to control mice (CTL) exposed only to normoxia (no treatment). An admittance catheter was used to measure pressure-volume loops at baseline and during vena cava occlusion, with mice ventilated with either room air or 8% oxygen, from which pulmonary hemodynamics, RV function, and ventricular-vascular coupling efficiency (ηvvc) were calculated. Proline analog treatment limited increases in RV afterload (neither effective arterial elastance Ea nor total pulmonary vascular resistance significantly increased compared to CTL with CHOP-PEG), limited the development of pulmonary hypertension (CHOP-PEG reduced right ventricular systolic pressure by 10% compared to HPH, p < 0.05), and limited RV hypertrophy (CHOP-PEG reduced RV mass by 18% compared to HPH, p < 0.005). In an acutely hypoxic state, treatment improved RV function (CHOP-PEG increased end-systolic elastance Ees by 43%, p < 0.05) and maintained ηvvc at control, room air levels. CHOP-PEG also decreased lung collagen content by 12% measured biochemically compared to HPH (p < 0.01), with differences evident in large and small pulmonary arteries by histology. Our results demonstrate that preventing new collagen synthesis limits pulmonary hypertension development by reducing collagen accumulation in the pulmonary arteries that affect RV afterload. In particular, the proline analog limited structural and functional changes in distal pulmonary arteries in this model of early and somewhat mild pulmonary hypertension. We conclude that collagen plays an important role in small pulmonary artery remodeling and, thereby, affects RV structure and function changes induced by chronic hypoxia.
Collapse
Affiliation(s)
- David Schreier
- Department of Biomedical Engineering, University of Wisconsin, 2145 ECB, 1550 Engineering Drive, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
33
|
Chen H, Hwang H, McKee LAK, Perez JN, Regan JA, Constantopoulos E, Lafleur B, Konhilas JP. Temporal and morphological impact of pressure overload in transgenic FHC mice. Front Physiol 2013; 4:205. [PMID: 23986715 PMCID: PMC3753457 DOI: 10.3389/fphys.2013.00205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/19/2013] [Indexed: 01/19/2023] Open
Abstract
Although familial hypertrophic cardiomyopathy (FHC) is characterized as cardiac disease in the absence of overt stressors, disease penetrance, and pathological progression largely depend on modifying factors. Accordingly, pressure overload by transverse aortic constriction (TAC) was induced in 2-month-old, male mice with and without a FHC (R403Q) mutation in α-myosin heavy chain. A significantly greater number of FHC mice (n = 8) than wild-type (WT) mice (n = 5) died during the 9-week study period. TAC induced a significant increase in cardiac mass whether measured at 2 or 9 weeks post-TAC in both WT and FHC mice, albeit to a different extent. However, the temporal and morphological trajectory of ventricular remodeling was impacted by the FHC transgene. Both WT and FHC hearts responded to TAC with an early (2 weeks post-TAC) and significant augmentation of the relative wall thickness (RWT) indicative of concentric hypertrophy. By 9 weeks post-TAC, RWT decreased in WT hearts (eccentric hypertrophy) but remained elevated in FHC hearts. WT hearts following TAC demonstrated enhanced cardiac function as measured by the end-systolic pressure-volume relationship, pre-load recruitable stroke work (PRSW), and myocardial relaxation indicative of compensatory hypertrophy. Similarly, TAC induced differential histological and cellular remodeling; TAC reduced expression of the sarcoplasmic reticulum Ca(2+)-ATPase (2a) (SERCA2a; 2 and 9 weeks) and phospholamban (PLN; 2 weeks) but increased PLN phosphorylation (2 weeks) and β-myosin heavy chain (β-MyHC; 9 weeks) in WT hearts. FHC-TAC hearts showed increased β-MyHC (2 and 9 weeks) and a late (9 weeks) decrease in PLN expression concomitant with a significant increase in PLN phosphorylation. We conclude that FHC hearts respond to TAC induced pressure overload with increased premature death, severe concentric hypertrophy, and a differential ability to undergo morphological, functional, or cellular remodeling compared to WT hearts.
Collapse
Affiliation(s)
- Hao Chen
- Molecular Cardiovascular Research Program, Department of Physiology, University of Arizona Tucson, AZ, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang W, McKinnie SMK, Patel VB, Haddad G, Wang Z, Zhabyeyev P, Das SK, Basu R, McLean B, Kandalam V, Penninger JM, Kassiri Z, Vederas JC, Murray AG, Oudit GY. Loss of Apelin exacerbates myocardial infarction adverse remodeling and ischemia-reperfusion injury: therapeutic potential of synthetic Apelin analogues. J Am Heart Assoc 2013; 2:e000249. [PMID: 23817469 PMCID: PMC3828798 DOI: 10.1161/jaha.113.000249] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [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 Coronary artery disease leading to myocardial ischemia is the most common cause of heart failure. Apelin (APLN), the endogenous peptide ligand of the APJ receptor, has emerged as a novel regulator of the cardiovascular system. METHODS AND RESULTS Here we show a critical role of APLN in myocardial infarction (MI) and ischemia-reperfusion (IR) injury in patients and animal models. Myocardial APLN levels were reduced in patients with ischemic heart failure. Loss of APLN increased MI-related mortality, infarct size, and inflammation with drastic reductions in prosurvival pathways resulting in greater systolic dysfunction and heart failure. APLN deficiency decreased vascular sprouting, impaired sprouting of human endothelial progenitor cells, and compromised in vivo myocardial angiogenesis. Lack of APLN enhanced susceptibility to ischemic injury and compromised functional recovery following ex vivo and in vivo IR injury. We designed and synthesized two novel APLN analogues resistant to angiotensin converting enzyme 2 cleavage and identified one analogue, which mimicked the function of APLN, to be markedly protective against ex vivo and in vivo myocardial IR injury linked to greater activation of survival pathways and promotion of angiogenesis. CONCLUSIONS APLN is a critical regulator of the myocardial response to infarction and ischemia and pharmacologically targeting this pathway is feasible and represents a new class of potential therapeutic agents.
Collapse
Affiliation(s)
- Wang Wang
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Karakikes I, Chaanine AH, Kang S, Mukete BN, Jeong D, Zhang S, Hajjar RJ, Lebeche D. Therapeutic cardiac-targeted delivery of miR-1 reverses pressure overload-induced cardiac hypertrophy and attenuates pathological remodeling. J Am Heart Assoc 2013; 2:e000078. [PMID: 23612897 PMCID: PMC3647279 DOI: 10.1161/jaha.113.000078] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background MicroRNAs (miRNAs) play a key role in the development of heart failure, and recent studies have shown that the muscle‐specific miR‐1 is a key regulator of cardiac hypertrophy. We tested the hypothesis that chronic restoration of miR‐1 gene expression in vivo will regress hypertrophy and protect against adverse cardiac remodeling induced by pressure overload. Methods and Results Cardiac hypertrophy was induced by left ventricular pressure overload in male Sprague‐Dawley rats subjected to ascending aortic stenosis. When the hypertrophy was established at 2 weeks after surgery, the animals were randomized to receive either an adeno‐associated virus expressing miR‐1 (AAV9.miR‐1) or green fluorescent protein (GFP) as control (AAV9.GFP) via a single‐bolus tail‐vein injection. Administration of miR‐1 regressed cardiac hypertrophy (left ventricular posterior wall thickness,; 2.32±0.08 versus 2.75±0.07 mm, P<0.001) and (left ventricular septum wall thickness, 2.23±0.06 versus 2.54±0.10 mm, P<0.05) and halted the disease progression compared with control‐treated animals, as assessed by echocardiography (fractional shortening, 37.60±5.01% versus 70.68±2.93%, P<0.05) and hemodynamic analyses (end‐systolic pressure volume relationship/effective arterial elastance, 1.87±0.46 versus 0.96±0.38, P<0.05) after 7 weeks of treatment. Additionally, miR‐1 replacement therapy lead to a marked reduction of myocardial fibrosis, an improvement in calcium handling, inhibition of apoptosis, and inactivation of the mitogen‐activated protein kinase signaling pathways, suggesting a favorable effect on preventing the maladaptive ventricular remodeling. We also identified and validated a novel bona fide target of miR‐1, Fibullin‐2 (Fbln2), a secreted protein implicated in extracellular matrix remodeling. Conclusions Taken together, our findings suggest that restoration of miR‐1 gene expression is a potential novel therapeutic strategy to reverse pressure‐induced cardiac hypertrophy and prevent maladaptive cardiac remodeling.
Collapse
Affiliation(s)
- Ioannis Karakikes
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kutty S, Kottam AT, Padiyath A, Bidasee KR, Li L, Gao S, Wu J, Lof J, Danford DA, Kuehne T. Validation of admittance computed left ventricular volumes against real-time three-dimensional echocardiography in the porcine heart. Exp Physiol 2013; 98:1092-101. [DOI: 10.1113/expphysiol.2012.070821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
37
|
Larson ER, Feldman MD, Valvano JW, Pearce JA. Analysis of the spatial sensitivity of conductance/admittance catheter ventricular volume estimation. IEEE Trans Biomed Eng 2013; 60:2316-24. [PMID: 23559022 DOI: 10.1109/tbme.2013.2256134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Conductance catheters are known to have a nonuniform spatial sensitivity due to the distribution of the electric field. The Geselowitz relation is applied to murine and multisegment conductance catheters using finite element models to determine the spatial sensitivity in a uniform medium and simplified left ventricle models. A new formulation is proposed that allows determination of the spatial sensitivity to admittance. Analysis of FEM numerical modeling results using the Geselowitz relation provides a true measure of parallel conductance in simplified left ventricle models for assessment of the admittance method and hypertonic saline techniques. The spatial sensitivity of blood conductance (Gb) is determined throughout the cardiac cycle. Gb is converted to volume using Wei's equation to determine if the presence of myocardium alters the nonlinear relationship through changes to the electric field. Results show that muscle conductance (Gm) from the admittance method matches results from the Geselowitz relation and that the relationship between Gb and volume is accurately fit using Wei's equation. Single-segment admittance measurements in large animals result in a more evenly distributed sensitivity to the LV blood pool. The hypertonic saline method overestimates parallel conductance throughout the cardiac cycle in both murine and multisegment conductance catheters.
Collapse
Affiliation(s)
- Erik R Larson
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX 78712, USA.
| | | | | | | |
Collapse
|
38
|
Chaanine AH, Gordon RE, Kohlbrenner E, Benard L, Jeong D, Hajjar RJ. Potential role of BNIP3 in cardiac remodeling, myocardial stiffness, and endoplasmic reticulum: mitochondrial calcium homeostasis in diastolic and systolic heart failure. Circ Heart Fail 2013; 6:572-83. [PMID: 23508759 DOI: 10.1161/circheartfailure.112.000200] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We have shown that BNIP3 expression is significantly increased in heart failure (HF). In this study, we tested the effects of BNIP3 manipulation in HF. METHODS AND RESULTS In a rat model of pressure overload HF, BNIP3 knockdown significantly decreased left ventricular (LV) volumes with significant improvement in LV diastolic and systolic function. There were significant decreases in myocardial apoptosis and LV interstitial fibrosis. Ultrastructurally, BNIP3 knockdown attenuated mitochondrial fragmentation and restored mitochondrial morphology and integrity. On the molecular level, there were significant decreases in endoplasmic reticulum (ER) stress and mitochondrial apoptotic markers. One of the mechanisms by which BNIP3 mediates mitochondrial dysfunction is via the oligomerization of the voltage-dependent anion channels causing a shift of calcium from the ER to mitochondrial compartments, leading to the decrease in ER calcium content, mitochondrial damage, apoptosis, and LV interstitial fibrosis, and hence contributes to both systolic and diastolic myocardial dysfunction, respectively. In systolic HF, the downregulation of SERCA2a (sarcoplasmic-endoplasmic reticulum calcium ATPase), along with an increased BNIP3 expression, further worsen myocardial diastolic and systolic function and contribute to the major remodeling seen in systolic HF as compared with diastolic HF with normal SERCA2a expression. CONCLUSIONS The increase in BNIP3 expression contributes mainly to myocardial diastolic dysfunction through mitochondrial apoptosis, LV interstitial fibrosis, and to some extent to myocardial systolic dysfunction attributable to the shift of calcium from the ER to the mitochondria and to the decrease in ER calcium content. However, SERCA2a downregulation remains a prerequisite for the major LV remodeling seen in systolic HF.
Collapse
Affiliation(s)
- Antoine H Chaanine
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
39
|
Clark JE, Marber MS. Advancements in pressure-volume catheter technology - stress remodelling after infarction. Exp Physiol 2012; 98:614-21. [PMID: 23064506 DOI: 10.1113/expphysiol.2012.064733] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microconductance catheters have been successfully applied to measure left ventricular (LV) function in the mouse to assess cardiac or pharmacological interventions for a number of years. New complex admittance methods produce an estimate of the parallel admittance of cardiac muscle that can be used to correct the measurement in real time. This contrasts with existing conductance technologies that require in vivo calibration using a bolus of hypertonic saline. Here, we report the application of this emerging technology in the context of myocardial infarction and LV remodelling. Using a combination of high-resolution ultrasound and LV conductance catheters, we compared measures of LV function using an admittance system and a traditional conductance-derived pressure-volume (PV) system. We subjected C57BL/6 mice to focal myocardial ischaemia-reperfusion by transient ligation of the left anterior descending coronary artery and assessed cardiac function with different systems to determine the reliability and accuracy of these methods to distinguish between normal and dysfunctional ventricle. We demonstrate that the admittance PV system, in our hands, provides a straightforward solution for assessing LV function in mice. Using this technique in combination with other established methods, we measured LV dysfunction following coronary artery occlusion and reperfusion, which can be ameliorated using a known preconditioning agent (CORM-3), and found that functional read-outs are representative of other methods. We have found that, especially in diseased tissue, LV pressure-volume loops derived from complex admittance provide a reproducible and reliable method of determining LV function without the need for technically challenging calibration. Our data suggest that admittance records accurate/physiological LV cavity volumes when compared with other invasive methods in the same animal. This emerging technology is both effective and reproducible for measuring LV function and dysfunction in the mouse, without the need for complicated interventions to calibrate the measurements or training in a new technology. This may mark the way towards a fast and accurate assessment of murine cardiac function in normal animals and disease models.
Collapse
Affiliation(s)
- James E Clark
- Department of Cardiology, King's College London, British Heart Foundation Excellence Centre, 4th Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.
| | | |
Collapse
|
40
|
Oh JG, Jeong D, Cha H, Kim JM, Lifirsu E, Kim J, Yang DK, Park CS, Kho C, Park S, Yoo YJ, Kim DH, Kim J, Hajjar RJ, Park WJ. PICOT increases cardiac contractility by inhibiting PKCζ activity. J Mol Cell Cardiol 2012; 53:53-63. [DOI: 10.1016/j.yjmcc.2012.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/06/2012] [Accepted: 03/09/2012] [Indexed: 11/28/2022]
|
41
|
Detombe SA, Xiang FL, Dunmore-Buyze J, White JA, Feng Q, Drangova M. Rapid microcomputed tomography suggests cardiac enlargement occurs during conductance catheter measurements in mice. J Appl Physiol (1985) 2012; 113:142-8. [PMID: 22518829 DOI: 10.1152/japplphysiol.00831.2011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Conductance catheters (CC) represent an established method of determining cardiac function in mice; however, the potentially detrimental effects a catheter may have on the mouse heart have never been evaluated. The present study takes advantage of rapid three-dimensional (3D) microcomputed tomography (CT) to compare simultaneously acquired micro-CT and CC measurements of left ventricular (LV) volumes in healthy and infarcted mice and to determine changes in LV volume and function associated with CC insertion. LV volumes were measured in C57BL/6 mice (10 healthy, 10 infarcted, 2% isoflurane anesthesia) using a 1.4-Fr Millar CC. 3D micro-CT images of each mouse were acquired before CC insertion as well as during catheterization. Each CT scan produced high-resolution images throughout the entire cardiac cycle in <1 min, enabling accurate volume measurements as well as direct visualization of the CC within the LV. Bland-Altman analysis demonstrated that CC measurements underestimate volume compared with CT measurements in both healthy [bias of -18.4 and -28.9 μl for end-systolic (ESV) and end-diastolic volume (EDV), respectively] and infarcted mice (ESV = -51.6 μl and EDV = -71.7 μl); underestimation was attributed to the off-center placement of the catheter. Individual evaluation of each heart revealed LV dilation following CC insertion in 40% of mice in each group. No change in ejection fraction was observed, suggesting the enlargement was caused by volume overload associated with disruption of the papillary muscles or chords. The enlargement witnessed was not significant; however, the results suggest the potential for CC insertion to detrimentally affect mouse myocardium, necessitating further investigation.
Collapse
Affiliation(s)
- Sarah A Detombe
- Imaging Research Laboratories, Robarts Research Institute, Western University, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
42
|
Kottam A, Dubois J, McElligott A, Henderson KK. Novel approach to admittance to volume conversion for ventricular volume measurement. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2011:2514-7. [PMID: 22254852 DOI: 10.1109/iembs.2011.6090696] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The conductance catheter is a widely used tool to determine ventricular volumes in animal models. A tetra-polar catheter is inserted into the ventricle to measure instantaneous conductance, which is a combination of ventricular blood and surrounding myocardium. Various techniques have been used to separate the blood conductance signal from the combined measured signal [1], [2]. The blood conductance is then converted to volume using a linear relationship proposed by Baan [1] or an improved non linear relationship proposed by Wei [3]. We propose a novel approach that uses the combined blood-muscle signal to calculate volume, thereby eliminating the need to subtract out the muscle. In vivo experiments were performed in mice to validate this new approach and the results were compared with volumes obtained using ultrasound imaging.
Collapse
Affiliation(s)
- Anil Kottam
- Department of Biomedical Engineering,University of Texas at Austin, Austin, TX 78712, USA.
| | | | | | | |
Collapse
|
43
|
Constantinides C, Angeli S, Kossivas F, Ktorides P. Underestimation of Murine Cardiac Hemodynamics Using Invasive Catheters: Errors, Limitations, and Remedies. Cardiovasc Eng Technol 2012. [DOI: 10.1007/s13239-012-0084-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
44
|
Martens A, Gruh I, Dimitroulis D, Rojas SV, Schmidt-Richter I, Rathert C, Khaladj N, Gawol A, Chikobava MG, Martin U, Haverich A, Kutschka I. Rhesus monkey cardiosphere-derived cells for myocardial restoration. Cytotherapy 2012; 13:864-72. [PMID: 21843109 DOI: 10.3109/14653249.2011.571247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Cardiosphere-derived cells (CDC) have been proposed as a promising myocardial stem cell source for cardiac repair. They have been isolated from human, porcine and rodent cardiac biopsies. However, their usefulness for myocardial restoration remains controversial. We aimed to determine the survival, differentiation and functional effects of Rhesus monkey CDC (RhCDC) in a mouse model of myocardial infarction. METHODS RhCDC were isolated and characterized by flow cytometry and reverse transcriptase (RT)-polymerase chain reaction (PCR) and compared with human CDC. They were injected intramyocardially into severe combined immune deficiency (SCID) beige mice after ligature of the left anterior descending artery (LAD). Phosphate-buffered saline (PBS) served as placebo. Medium treatment alone was used to distinguish between cellular and non-cellular effects. Animals were divided into a non-infarcted control group (n = 7), infarct control groups (n = 24), medium-treated infarct groups (n = 35) and RhCDC-treated infarct groups (n = 33). Follow-up was either 1 or 4 weeks. LV function was assessed by pressure-volume loop analysis. Differentiation was analyzed by immunhistochemical profiling and RT-PCR. RESULTS Proliferating RhCDC grafts were detected after transplantation in an acute infarct model. RhCDC as well as medium treatment protected myocardium within the infarct area and improved LV function. RhCDC had a superior regenerative effect than medium alone. CONCLUSIONS For the first time, RhCDC have been used for the restoration of infarcted myocardium. RhCDC proliferated in vivo and positively influenced myocardial remodeling. This effect could be mimicked by treatment with unconditioned medium alone, emphasizing a non-cellular paracrine therapeutic mechanism. However, as a robust cardiac stem cell source, CDC might be useful to evoke prolonged paracrine actions in cardiac stem cell therapy.
Collapse
Affiliation(s)
- Andreas Martens
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chaanine AH, Jeong D, Liang L, Chemaly ER, Fish K, Gordon RE, Hajjar RJ. JNK modulates FOXO3a for the expression of the mitochondrial death and mitophagy marker BNIP3 in pathological hypertrophy and in heart failure. Cell Death Dis 2012; 3:265. [PMID: 22297293 PMCID: PMC3288347 DOI: 10.1038/cddis.2012.5] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bcl-2 E1B 19-KDa interacting protein 3 (BNIP3) is a mitochondrial death and mitophagy marker, which is involved in inducing cardiac remodeling post myocardial infarction. In this study, we show that BNIP3 expression increases in stressed cardiomyocytes in vitro and in response to pressure overload in vivo, and that its transcription is directly related to JNK activity. BNIP3 expression gradually increased in the first weeks after pressure overload and peaked at the heart failure stage. Ultrastructurally, the mitochondrial area was inversely proportional to BNIP3 expression. Both JNK and AKT activities increased with pressure overload; however, JNK signaling dominated over AKT signaling for the activation of the transcription factor FOXO3a and for the transcription of its effector, BNIP3. 3-methyladenine attenuated JNK signaling and significantly decreased BNIP3 expression and reversed cardiac remodeling in heart failure. Ultrastructurally, the mitochondrial area was significantly increased in the 3-methyladenine group compared with placebo. Moreover, adenoviral gene delivery of dominant negative JNK in a rat model of pressure overload hypertrophy abolished the increase in BNIP3 expression in response to pressure overload. These results suggest that JNK signaling is a critical modulator of the transcription factor FOXO3a driving the expression of its effector, BNIP3, in heart failure and that JNK, through BNIP3, induces mitochondrial apoptosis and mitophagy.
Collapse
Affiliation(s)
- A H Chaanine
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Larson ER, Pearce JA. Application of the Geselowitz relationship to the murine conductance catheter. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2012:3247-3250. [PMID: 23366618 DOI: 10.1109/embc.2012.6346657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Conductance catheters are known to have a nonuniform spatial sensitivity due to the distribution of the electric field. The Geselowitz relation is applied to the murine conductance catheter using a finite element model to determine catheter's spatial sensitivity in uniform media. Further analysis of FEM numerical modeling results using the Geselowitz relation provides a true measure of parallel conductance in a simplified murine left ventricle for assessment of the admittance method and hypertonic saline techniques. The spatial sensitivity of blood conductance (G(b)) is determined throughout the cardiac cycle. G(b) is converted to volume using Wei's equation to determine if the presence of myocardium alters the nonlinear relationship through changes to the electric field shape. Results show that the admittance method correctly calculates G(b) in comparison to the Geselowitz relation, and that the relationship between G(b) and volume is accurately fit using Wei's equation.
Collapse
Affiliation(s)
- Erik R Larson
- University of Texas at Austin, 1 University Station, Austin, TX 78712, USA
| | | |
Collapse
|
47
|
Cingolani OH, Kass DA. Pressure-volume relation analysis of mouse ventricular function. Am J Physiol Heart Circ Physiol 2011; 301:H2198-206. [PMID: 21926344 DOI: 10.1152/ajpheart.00781.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nearly 40 years ago, the Sagawa laboratory spawned a renaissance in the use of instantaneous ventricular pressure-volume (P-V) relations to assess cardiac function. Since then, this analysis has taken hold as the most comprehensive way to quantify ventricular chamber function and energetics and cardiovascular interactions. First studied in large mammalian hearts and later in humans employing a catheter-based method, P-V analysis was translated to small rodents in the late 1990s by the Kass laboratory. Over the past decade, this approach has become a gold standard for comprehensive examination of in vivo cardiac function in mice, facilitating a new era of molecular cardiac physiology. The catheter-based method remains the most widely used approach in mice. In this brief review, we discuss this instrumentation, the theory behind its use, and how volume signals are calibrated and discuss elements of P-V analysis. The goal is to provide a convenient summary of earlier investigations and insights for users whose primary interests lie in genetic/molecular studies rather than in biomedical engineering.
Collapse
Affiliation(s)
- Oscar H Cingolani
- Division of Cardiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
48
|
Kho C, Lee A, Jeong D, Oh JG, Chaanine AH, Kizana E, Park WJ, Hajjar RJ. SUMO1-dependent modulation of SERCA2a in heart failure. Nature 2011; 477:601-5. [PMID: 21900893 PMCID: PMC3443490 DOI: 10.1038/nature10407] [Citation(s) in RCA: 278] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 08/01/2011] [Indexed: 12/23/2022]
Abstract
SR Ca2+ ATPase 2a (SERCA2a) is a critical ATPase responsible for Ca2+ re-uptake during excitation-contraction coupling. Impaired SR Ca2+ uptake resulting from decreased expression and reduced activity of SERCA2a is a hallmark of heart failure (HF)1. Accordingly, restoration of SERCA2a expression by gene transfer has proven to be effective in improving cardiac function in HF patients2 as well as in animal models3. The small ubiquitin-related modifier (SUMO) can be conjugated to lysine residues of target proteins4, which is involved in most cellular process5. Here, we show that SERCA2a is SUMOylated at lysine 480 and 585 and that this SUMOylation is essential for preserving SERCA2a ATPase activity and stability. The levels of SUMO1 and SUMOylation of SERCA2a itself were greatly reduced in failing hearts. SUMO1 restitution by adeno-associated virus-mediated gene delivery maintained protein abundance of SERCA2a and significantly improved cardiac function in HF mice. This effect was comparable to SERCA2a gene delivery. Moreover, SUMO1 overexpression in isolated cardiomyocytes augmented contractility and accelerated Ca2+ decay. Transgene-mediated SUMO1 overexpression rescued pressure overload-induced cardiac dysfunction concomitantly with increased SERCA2a function. By contrast, down-regulation of SUMO1 using shRNA accelerated pressure overload-induced deterioration of cardiac function and was accompanied by decreased SERCA2a function. However, knockdown of SERCA2a resulted in severe contractile dysfunction both in vitro and in vivo, which was not rescued by overexpression of SUMO1. Taken together, our data show that SUMOylation is a critical post-translational modification that regulates SERCA2a function and provides a platform for the design of novel therapeutic strategies for HF.
Collapse
Affiliation(s)
- Changwon Kho
- Cardiovascular Research Center, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, Box 1030, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Constantinides C, Angeli SI, Mean RJ. Murine cardiac catheterizations and hemodynamics: on the issue of parallel conductance. IEEE Trans Biomed Eng 2011; 58:3260-8. [PMID: 21900070 DOI: 10.1109/tbme.2011.2167147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Catheter-based measurements are extensively used nowadays in animal models to quantify global left ventricular (LV) cardiac function and hemodynamics. Conductance catheter measurements yield estimates of LV volumes. Such estimates, however, are confounded by the catheter's nonhomogeneous emission field and the contribution to the total conductance of surrounding tissue or blood conductance values (other than LV blood), a term often known as parallel conductance. In practice, in most studies, volume estimates are based on the assumptions that the catheter's electric field is homogeneous and that parallel conductance is constant, despite prior results showing that these assumptions are incorrect. This study challenges the assumption for spatial homogeneity of electric field excitation of miniature catheters and investigated the electric field distribution of miniature catheters in the murine heart, based on cardiac model-driven (geometric, lump component) simulations and noninvasive imaging, at both systolic and diastolic cardiac phases. Results confirm the nonuniform catheter emission field, confined spatially within the LV cavity and myocardium, falling to 10% of its peak value at the ring electrode surface, within 1.1-2.0 mm, given a relative tissue permittivity of 33,615. Additionally, <1% of power leaks were observed into surrounding cavities or organs at end-diastole. Temporally varying parallel conductance effects are also confirmed, becoming more prominent at end-systole.
Collapse
|
50
|
Constantinides C, Angeli S, Mean R. Murine cardiac hemodynamics following manganese administration under isoflurane anesthesia. Ann Biomed Eng 2011; 39:2706-20. [PMID: 21818535 DOI: 10.1007/s10439-011-0367-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/21/2011] [Indexed: 12/22/2022]
Abstract
This study examines (a) the temporal stability of hemodynamic indices of systolic and diastolic function in C57BL/6 mice under 1.5% isoflurane (ISO) (v/v) anesthesia conditions in 50:50 O(2)/N(2)O (v/v) within 90 min post-induction, and (b) the effects of Mn(2+) on the mouse hemodynamic response in male C57BL/6 mice (n = 16). Left ventricular catheterizations allowed estimation of the hemodynamic indices. Hypertonic saline infusion (10%) allowed absolute volume quantification in conjunction with a separate series of aortic flow experiments (n = 3). In a separate cohort of mice (n = 6), MnCl(2) (190 nmoles/g/bw) was infused via the left jugular for 29-39 min, following 11 min of baseline recording, to assess temporal responses. Stable temporal hemodynamic responses were achieved in control mice under ISO anesthesia. Hemodynamic indices during control, time-matched-control, baseline-Mn, and Mn-infused periods, were within normal expected ranges. No chronotropic changes were observed. Significant differences in systolic and diastolic cardiac indices of function (HR, EF, ESP, dP/dt (max), dP/dt (min), PAMP, τ(glantz), and τ(weiss)) resulted between baseline-Mn and Mn-infused time periods in Mn-treated mice at the 1% significance (p < 0.001). Transient positive, or negative, or positive followed by negative evoked pressure-volume loop shifts were observed (exemplified through changes in the end-systolic pressure-volume relationship and dP/dt (max)) in Mn-infusion studies. It is concluded that Mn(2+) can be used safely for prolonged mouse imaging studies, however, the significant variations elicited in cardiovascular hemodynamics post-manganese infusion, necessitate further investigations for its suitability and appropriateness for quantification of global cardiac function in image-based phenotyping.
Collapse
Affiliation(s)
- C Constantinides
- Laboratory of Physiology and Biomedical Imaging, Department of Mechanical and Manufacturing Engineering, School of Engineering, University of Cyprus, Nicosia, Cyprus.
| | | | | |
Collapse
|