1
|
Bracamonte JH, Watkins L, Pat B, Dell’Italia LJ, Saucerman JJ, Holmes JW. Contributions of mechanical loading and hormonal changes to eccentric hypertrophy during volume overload: A Bayesian analysis using logic-based network models. PLoS Comput Biol 2025; 21:e1012390. [PMID: 40238825 PMCID: PMC12040246 DOI: 10.1371/journal.pcbi.1012390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 04/29/2025] [Accepted: 02/23/2025] [Indexed: 04/18/2025] Open
Abstract
Primary mitral regurgitation (MR) is a pathology that alters mechanical loading on the left ventricle, triggers an array of compensatory neurohormonal responses, and induces a distinctive ventricular remodeling response known as eccentric hypertrophy. Drug therapies may alleviate symptoms, but only mitral valve repair or replacement can provide significant recovery of cardiac function and dimensions. Questions remain about the optimal timing of surgery, with 20% of patients developing systolic dysfunction post-operatively despite being treated according to the current guidelines. Thus, better understanding of the hypertrophic process in the setting of ventricular volume overload (VO) is needed to improve and better personalize the management of MR. To address this knowledge gap, we employ a Bayesian approach to combine data from 70 studies on experimental volume overload in dogs and rats and use it to calibrate a logic-based network model of hypertrophic signaling in myocytes. The calibrated model predicts that growth in experimental VO is mostly driven by the neurohormonal response, with an initial increase in myocardial tissue stretch being compensated by subsequent remodeling fairly early in the time course of VO. This observation contrasts with a common perception that volume-overload hypertrophy is driven primarily by increased myocyte strain. The model reproduces many aspects of 43 studies not used in its calibration, including infusion of individual hypertrophic agonists alone or in combination with various drugs commonly employed to treat heart failure, as well as administration of some of those drugs in the setting of experimental volume overload. We believe this represents a promising approach to using the known structure of an intracellular signaling network to integrate information from multiple studies into quantitative predictions of the range of expected responses to potential interventions in the complex setting of cardiac hypertrophy driven by a combination of hormonal and mechanical factors.
Collapse
Affiliation(s)
- Johane H. Bracamonte
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lionel Watkins
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Betty Pat
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J. Dell’Italia
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey W. Holmes
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
2
|
Mueller KMA, Mansi S, De-Juan-Pardo EM, Mela P. Advances in melt electrowriting for cardiovascular applications. Front Bioeng Biotechnol 2024; 12:1425073. [PMID: 39355277 PMCID: PMC11442423 DOI: 10.3389/fbioe.2024.1425073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Melt electrowriting (MEW) is an electric-field-assisted additive biofabrication technique that has brought significant advancements to bioinspired scaffold design for soft tissue engineering and beyond. Owing to its targeted microfiber placement, MEW has become a powerful platform technology for the fabrication of in vitro disease models up to functional biohybrid constructs that are investigated in vivo to reach clinical translation soon. This work provides a concise overview of this rapidly evolving field by highlighting the key contributions of MEW to cardiovascular tissue engineering. Specifically, we i) pinpoint the methods to introduce microvascular networks in thick 3D constructs benefitting from (sacrificial) MEW microfibers, ii) report MEW-based concepts for small-diameter vascular grafts and stents, iii) showcase how contracting cardiac tissues can profit from the tunable structure-property relationship of MEW scaffolds, and iv) address how complete regenerative heart valves can be built on complex fiber scaffold architectures that recapitulate J-shaped tensile properties and tissue heterogeneity. Lastly, we touch on novel biomaterial advancements and discuss the technological challenges of MEW to unlock the full potential of this transformative technology.
Collapse
Affiliation(s)
- Kilian Maria Arthur Mueller
- Technical University of Munich, TUM School of Engineering and Design, Department of Mechanical Engineering, Chair of Medical Materials and Implants, Munich Institute of Biomedical Engineering (MIBE), Munich Institute of Integrated Materials, Energy and Process Engineering (MEP), Munich, Germany
| | - Salma Mansi
- Technical University of Munich, TUM School of Engineering and Design, Department of Mechanical Engineering, Chair of Medical Materials and Implants, Munich Institute of Biomedical Engineering (MIBE), Munich Institute of Integrated Materials, Energy and Process Engineering (MEP), Munich, Germany
| | - Elena M De-Juan-Pardo
- T3mPLATE, Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre and University of Western Australia Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- School of Engineering, The University of Western Australia, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
| | - Petra Mela
- Technical University of Munich, TUM School of Engineering and Design, Department of Mechanical Engineering, Chair of Medical Materials and Implants, Munich Institute of Biomedical Engineering (MIBE), Munich Institute of Integrated Materials, Energy and Process Engineering (MEP), Munich, Germany
| |
Collapse
|
3
|
Kawai A, Nagatomo Y, Yukino-Iwashita M, Ikegami Y, Takei M, Goda A, Kohno T, Mizuno A, Kitamura M, Nakano S, Sakamoto M, Shiraishi Y, Kohsaka S, Adachi T, Yoshikawa T. Supra-normal and mildly reduced ejection fraction in women -An overlooked vulnerable subpopulation in heart failure. Int J Cardiol 2024; 409:132166. [PMID: 38744340 DOI: 10.1016/j.ijcard.2024.132166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/04/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Recently, patients with supra-normal left ventricular ejection fraction (snEF) are reported to have high risk of adverse outcomes, especially in women. We sought to evaluate sex-related differences in the association between LVEF and long-term outcomes in heart failure (HF) patients. METHODS The multicenter WET-HF Registry enrolled all patients hospitalized for acute decompensated HF (ADHF). We analyzed 3943 patients (age 77 years; 40.1% female) registered from 2006 to 2017. According to LVEF the patients were divided into the 3 groups: HF with reduced EF (HFrEF), mildly reduced EF (HFmrEF) and preserved EF. The primary endpoint was defined as the composite of cardiac death and ADHF rehospitalization after discharge. RESULTS In HFmrEF, implementation of guideline-directed medical therapy (GDMT) such as the combination of renin-angiotensin-system inhibitor (RASi) and β-blocker at discharge was significantly lower in women than men even after adjustment for covariates (p = 0.007). There were no such sex-related differences in HFrEF. Female sex was associated with higher incidence of the primary endpoint and ADHF rehospitalization after adjustment for covariates exclusively in HFmrEF. Restricted cubic spline analysis demonstrated a U-shaped relationship between LVEF and the hazard ratio of the primary endpoint showing higher event rate in HFmrEF and HFsnEF in women, but such relationship was not observed in men (p for interaction = 0.037). CONCLUSIONS In women, mrEF and snEF were associated with worse long-term outcomes. Additionally, sex-related differences in the GDMT implementation for HFmrEF highlight the need for further exploration, which might lead to creation of sex-specific guidelines to optimize HF management.
Collapse
Affiliation(s)
- Akane Kawai
- Department of Cardiology, National Defense Medical College, Tokorozawa, Japan
| | - Yuji Nagatomo
- Department of Cardiology, National Defense Medical College, Tokorozawa, Japan; Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan.
| | | | - Yukinori Ikegami
- Department of Cardiology, National Defense Medical College, Tokorozawa, Japan
| | - Makoto Takei
- Department of Cardiology, Saiseikai Central Hospital, Tokyo, Japan
| | - Ayumi Goda
- Department of Cardiovascular Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Takashi Kohno
- Department of Cardiovascular Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Atsushi Mizuno
- Department of Cardiology, St. Luke's International Hospital, Tokyo, Japan
| | | | - Shintaro Nakano
- Department of Cardiology, Saitama Medical University, International Medical Center, Hidaka, Japan
| | - Munehisa Sakamoto
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Yasuyuki Shiraishi
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Adachi
- Department of Cardiology, National Defense Medical College, Tokorozawa, Japan
| | | |
Collapse
|
4
|
Ohte N, Kikuchi S, Iwahashi N, Kinugasa Y, Dohi K, Takase H, Masai K, Inoue K, Okumura T, Hachiya K, Kitada S, Seo Y. Distinctive left ventricular-arterial and right ventricular-pulmonary arterial coupling observed in patients with heart failure and a higher left ventricular ejection fraction range. Eur Heart J Cardiovasc Imaging 2024; 25:774-781. [PMID: 38284416 DOI: 10.1093/ehjci/jeae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/23/2023] [Accepted: 01/23/2024] [Indexed: 01/30/2024] Open
Abstract
AIMS Higher left ventricular (LV) ejection fraction (EF) is related to unfavourable prognosis in patients with heart failure (HF) with preserved ejection fraction (HFpEF). The cause of this finding needs to be haemodynamically explained. Thus, we investigated this crucial issue from the perspective of LV-arterial (A) and right ventricular (RV)-pulmonary arterial (PA) coupling. METHODS AND RESULTS Study patients were derived from our prospective cohort study of patients hospitalized due to acute decompensated HF and LVEF > 40%. We divided the 255 patients into three groups: HF with mildly reduced EF (HFmrEF), HFpEF with 50% ≤ LVEF < 60%, and HFpEF with LVEF ≥ 60%. We compared LV end-systolic elastance (Ees), effective arterial elastance (Ea), and Ees/Ea as a representative of LV-A coupling among groups and compared the ratio of tricuspid annular plane excursion to peak pulmonary arterial systolic pressure (TAPSE/PASP) as a representative of RV-PA coupling. All-cause death and readmission due to HF-free survival was worse in the group with a higher LVEF range. Ees/Ea was greater in HFpEF patients with LVEF ≥ 60% (2.12 ± 0.57) than in those with 50% ≤ LVEF < 60% (1.20 ± 0.14) and those with HFmrEF (0.82 ± 0.09) (P < 0.001). PASP was increased in the groups with higher LVEF; however, TAPSE/PASP did not differ among groups (n = 168, P = 0.17). In a multivariate Cox proportional hazard model, TAPSE/PASP but not PASP was significantly related to event-free survival independent of LVEF. CONCLUSION HFpEF patients with higher LVEF have unfavourable prognosis and distinctive LV-A coupling: Ees/Ea is elevated up to 2.0 or more. Impaired RV-PA coupling also worsens prognosis in such patients. CLINICAL TRIAL REGISTRATION URL: https://www.umin.ac.jp/ctr/index.htm Unique identifier: UMIN000017725.
Collapse
Affiliation(s)
- Nobuyuki Ohte
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Shohei Kikuchi
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Noriaki Iwahashi
- Cardiovascular Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yoshiharu Kinugasa
- Department of Cardiovascular Medicine and Endocrinology and Metabolism, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroyuki Takase
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Kumiko Masai
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Katsuji Inoue
- Department of Cardiology, Pulmonology, Hypertension & Nephrology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Hachiya
- Department of Cardiology, Nagoya City University East Medical Center, Nagoya, Japan
| | - Shuichi Kitada
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yoshihiro Seo
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
5
|
Kawai A, Nagatomo Y, Yukino-Iwashita M, Nakazawa R, Yumita Y, Taruoka A, Takefuji A, Yasuda R, Toya T, Ikegami Y, Masaki N, Adachi T. Sex Differences in Cardiac and Clinical Phenotypes and Their Relation to Outcomes in Patients with Heart Failure. J Pers Med 2024; 14:201. [PMID: 38392634 PMCID: PMC10890585 DOI: 10.3390/jpm14020201] [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: 12/29/2023] [Revised: 01/22/2024] [Accepted: 02/11/2024] [Indexed: 02/24/2024] Open
Abstract
Biological sex is one of the major factors characterizing the heart failure (HF) patient phenotype. Understanding sex-related differences in HF is crucial to implement personalized care for HF patients with various phenotypes. There are sex differences in left ventricular (LV) remodeling patterns in the HF setting, namely, more likely concentric remodeling and diastolic dysfunction in women and eccentric remodeling and systolic dysfunction in men. Recently supra-normal EF (snLVEF) has been recognized as a risk of worse outcome. This pathology might be more relevant in female patients. The possible mechanism may be through coronary microvascular dysfunction and sympathetic nerve overactivation from the findings of previous studies. Further, estrogen deficit might play a significant role in this pathophysiology. The sex difference in body composition may also be related to the difference in LV remodeling and outcome. Lower implementation in guideline-directed medical therapy (GDMT) in female HFrEF patients might also be one of the factors related to sex differences in relation to outcomes. In this review, we will discuss the sex differences in cardiac and clinical phenotypes and their relation to outcomes in HF patients and further discuss how to provide appropriate treatment strategies for female patients.
Collapse
Affiliation(s)
- Akane Kawai
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yuji Nagatomo
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | | | - Ryota Nakazawa
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yusuke Yumita
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Akira Taruoka
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Asako Takefuji
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Risako Yasuda
- Department of Intensive Care, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Takumi Toya
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yukinori Ikegami
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Nobuyuki Masaki
- Department of Intensive Care, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Takeshi Adachi
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| |
Collapse
|
6
|
Zhang Y, Kalhöfer-Köchling M, Bodenschatz E, Wang Y. Physical model of end-diastolic and end-systolic pressure-volume relationships of a heart. Front Physiol 2023; 14:1195502. [PMID: 37670768 PMCID: PMC10475591 DOI: 10.3389/fphys.2023.1195502] [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: 03/28/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Left ventricular stiffness and contractility, characterized by the end-diastolic pressure-volume relationship (EDPVR) and the end-systolic pressure-volume relationship (ESPVR), are two important indicators of the performance of the human heart. Although much research has been conducted on EDPVR and ESPVR, no model with physically interpretable parameters combining both relationships has been presented, thereby impairing the understanding of cardiac physiology and pathology. Here, we present a model that evaluates both EDPVR and ESPVR with physical interpretations of the parameters in a unified framework. Our physics-based model fits the available experimental data and in silico results very well and outperforms existing models. With prescribed parameters, the new model is used to predict the pressure-volume relationships of the left ventricle. Our model provides a deeper understanding of cardiac mechanics and thus will have applications in cardiac research and clinical medicine.
Collapse
Affiliation(s)
- Yunxiao Zhang
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Moritz Kalhöfer-Köchling
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Eberhard Bodenschatz
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Institute for Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
- Laboratory of Atomic and Solid-State Physics and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Yong Wang
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
7
|
Ohte N, Kikuchi S, Iwahashi N, Kinugasa Y, Dohi K, Takase H, Masai K, Inoue K, Okumura T, Hachiya K, Kitada S, Seo Y. Unfavourable outcomes in patients with heart failure with higher preserved left ventricular ejection fraction. Eur Heart J Cardiovasc Imaging 2023; 24:293-300. [PMID: 36464890 DOI: 10.1093/ehjci/jeac240] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Newly introduced drugs for heart failure (HF) have been reported to improve the prognosis of HF with preserved ejection fraction (HFpEF) in the lower range of left ventricular ejection fraction (LVEF). We hypothesized that a higher LVEF is related to an unfavourable prognosis in patients with HFpEF. METHODS AND RESULTS We tested this hypothesis by analysing the data from a prospective multicentre cohort study in 255 patients admitted to the hospital due to decompensated HF (LVEF > 40% at discharge). The primary endpoint of this study was a composite outcome of all-cause death and readmission due to HF, and the secondary endpoint was readmission due to HF. LVEF and the mitral E/e' ratio were measured using echocardiography. In multicovariate parametric survival time analysis, LVEF [hazard ratio (HR) = 1.046 per 1% increase, P = 0.001], concurrent atrial fibrillation (AF) (HR = 3.203, P < 0.001), and E/e' (HR = 1.083 per 1.0 increase, P < 0.001) were significantly correlated with the primary endpoint. In addition to these covariates, angiotensin-converting enzyme inhibitor (ACEI)/angiotensin receptor blocker (ARB) use was significantly correlated with the secondary endpoint (HR = 0.451, P = 0.008). Diagnostic performance plot analysis demonstrated that the discrimination threshold value for LVEF that could identify patients prone to reaching the primary endpoint was ≥57.2%. The prevalence of AF or E/e' ratio did not differ significantly between patients with LVEF ≥ 58% and with 40% < LVEF < 58%. CONCLUSION A higher LVEF is independently related to poor prognosis in patients with HFpEF, in addition to concurrent AF and an elevated E/e' ratio. ACEI/ARB use, in contrast, was associated with improved prognosis, especially with regard to readmission due to HF. CLINICAL TRIAL REGISTRATION https://www.umin.ac.jp/ctr/index.htm. UNIQUE IDENTIFIER UMIN000017725.
Collapse
Affiliation(s)
- Nobuyuki Ohte
- Department of Cardiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Shohei Kikuchi
- Department of Cardiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Noriaki Iwahashi
- Cardiovascular Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yoshiharu Kinugasa
- Department of Cardiovascular Medicine and Endocrinology and Metabolism, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hiroyuki Takase
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Kumiko Masai
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Katsuji Inoue
- Department of Cardiology, Pulmonology, Hypertension & Nephrology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Takahiro Okumura
- Department of Cardiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kenta Hachiya
- Department of Cardiology, Nagoya City University East Medical Center, Nagoya, Japan
| | - Shuichi Kitada
- Department of Cardiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Yoshihiro Seo
- Department of Cardiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| |
Collapse
|
8
|
Furlani F, Campodoni E, Sangiorgi N, Montesi M, Sanson A, Sandri M, Panseri S. Electroconductive scaffolds based on gelatin and PEDOT:PSS for cardiac regeneration. Int J Biol Macromol 2022; 224:266-280. [DOI: 10.1016/j.ijbiomac.2022.10.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
|
9
|
Giao DM, Wang Y, Rojas R, Takaba K, Badathala A, Spaulding KA, Soon G, Zhang Y, Wang VY, Haraldsson H, Liu J, Saloner D, Guccione JM, Ge L, Wallace AW, Ratcliffe MB. Left ventricular geometry during unloading and the end-systolic pressure volume relationship: Measurement with a modified real-time MRI-based method in normal sheep. PLoS One 2020; 15:e0234896. [PMID: 32569290 PMCID: PMC7307770 DOI: 10.1371/journal.pone.0234896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 06/04/2020] [Indexed: 01/08/2023] Open
Abstract
The left ventricular (LV) end-systolic (ES) pressure volume relationship (ESPVR) is the cornerstone of systolic LV function analysis. We describe a 2D real-time (RT) MRI-based method (RTPVR) with separate software tools for 1) semi-automatic level set-based shape prior method (LSSPM) of the LV, 2) generation of synchronized pressure area loops and 3) calculation of the ESPVR. We used the RTPVR method to measure ventricular geometry, ES pressure area relationship (ESPAR) and ESPVR during vena cava occlusion (VCO) in normal sheep. 14 adult sheep were anesthetized and underwent measurement of LV systolic function. Ten of the 14 sheep underwent RTMRI and eight of the 14 underwent measurement with conductance catheter; 4 had both RTMRI and conductance measurements. 2D cross sectional RTMRI were performed at apex, mid-ventricle and base levels during separate VCOs. The Dice similarity coefficient was used to compare LSSPM and manual image segmentation and thus determine LSSPM accuracy. LV cross-sectional area, major and minor axis length, axis ratio, major axis orientation angle and ESPAR were measured at each LV level. ESPVR was calculated with a trapezoidal rule. The Dice similarity coefficient between LSSPM and manual segmentation by two readers was 87.31±2.51% and 88.13±3.43%. All cross sections became more elliptical during VCO. The major axis orientation shifted during VCO but remained in the septo-lateral direction. LV chamber obliteration at the apical level occurred during VCO in 7 of 10 sheep that underwent RTMRI. ESPAR was non-linear at all levels. Finally, ESPVR was non-linear because of apical collapse. ESPVR measured by conductance catheter (EES,Index = 2.23±0.66 mmHg/ml/m2) and RT (EES,Index = 2.31±0.31 mmHg/ml/m2) was not significantly different. LSSPM segmentation of 2D RT MRI images is accurate and allows calculation of LV geometry, ESPAR and ESPVR during VCO. In the future, RTPVR will facilitate determination of regional systolic material parameters underlying ESPVR.
Collapse
Affiliation(s)
- Duc M. Giao
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Yan Wang
- Department of Radiology, University of California, San Francisco, CA, United States of America
| | - Renan Rojas
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Kiyoaki Takaba
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Anusha Badathala
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Kimberly A. Spaulding
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Gilbert Soon
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Yue Zhang
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Vicky Y. Wang
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Henrik Haraldsson
- Department of Radiology, University of California, San Francisco, CA, United States of America
| | - Jing Liu
- Department of Radiology, University of California, San Francisco, CA, United States of America
| | - David Saloner
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Radiology, University of California, San Francisco, CA, United States of America
| | - Julius M. Guccione
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Liang Ge
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
| | - Arthur W. Wallace
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Anesthesia, University of California, San Francisco, CA, United States of America
| | - Mark B. Ratcliffe
- Veterans Affairs Medical Center, San Francisco, California, United States of America
- Department of Bioengineering, University of California, San Francisco, CA, United States of America
- Department of Surgery, University of California, San Francisco, CA, United States of America
- * E-mail:
| |
Collapse
|
10
|
A new non-invasive index for prognosis evaluation in patients with aortic stenosis. Sci Rep 2020; 10:7333. [PMID: 32355310 PMCID: PMC7193646 DOI: 10.1038/s41598-020-63777-z] [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: 11/30/2019] [Accepted: 03/31/2020] [Indexed: 11/09/2022] Open
Abstract
The global left ventricular (LV) contractility index, dσ*/dtmax measures the maximal rate of change in pressure-normalized LV wall stress. We aim to describe the trend of dσ*/dtmax in differing severity of aortic stenosis (AS) with preserved left ventricular ejection fraction (LVEF) and the association of dσ*/dtmax with clinical outcomes in moderate AS and severe AS. We retrospectively studied a total of 1738 patients with AS (550 mild AS, 738 moderate AS, 450 severe AS) and preserved LVEF ≥ 50% diagnosed from 1st January 2001 to 31st December 2015. dσ*/dtmax worsened with increasing severity of AS despite preserved LVEF (mild AS: 3.69 ± 1.28 s-1, moderate AS: 3.17 ± 1.09 s-1, severe AS: 2.58 ± 0.83 s-1, p < 0.001). Low dσ*/dtmax < 2.8 s-1 was independently associated with a higher composite outcome of aortic valve replacement, congestive cardiac failure admissions and all-cause mortality (adjusted hazard ratio 1.48, 95% CI: 1.25-1.77, p < 0.001). In conclusion, dσ*/dtmax declined with worsening AS despite preserved LVEF. Low dσ*/dtmax < 2.8 s-1 was independently associated with adverse clinical outcomes in moderate AS and severe AS with preserved LVEF.
Collapse
|
11
|
Kamimura D, Suzuki T, Hall ME, Wang W, Winniford MD, Shah AM, Rodriguez CJ, Butler KR, Mosley TH. Diastolic wall strain is associated with incident heart failure in African Americans: Insights from the atherosclerosis risk in communities study. J Cardiol 2018; 71:477-483. [PMID: 29203080 PMCID: PMC6100746 DOI: 10.1016/j.jjcc.2017.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Increased left ventricular (LV) myocardial stiffness may be associated with impaired LV hemodynamics and incident heart failure (HF). However, an indicator that estimates LV myocardial stiffness easily and non-invasively is lacking. The purpose of this study was to determine whether diastolic wall strain (DWS), an echocardiographic estimator of LV myocardial stiffness, is associated with incident HF in a middle-aged community-based cohort of African Americans. METHODS AND RESULTS We investigated associations between DWS and incident HF among 1528 African Americans (mean age 58.5 years, 66% women) with preserved LV ejection fraction (EF ≥50%) and without a history of cardiovascular disease in the Atherosclerosis Risk in Communities Study. Participants with the smallest DWS quintile (more LV myocardial stiffness) had a higher LV mass index, higher relative wall thickness, and lower arterial compliance than those in the larger four DWS quintiles (p<0.01 for all). Over a mean follow-up of 15.6 years, there were 251 incident HF events (incidence rate: 10.9 per 1000 person-years). After adjustment for traditional risk factors and incident coronary artery disease, both continuous and categorical DWS were independently associated with incident HF (HR 1.21, 95%CI 1.04-1.41 for 0.1 decrease in continuous DWS, p=0.014, HR 1.40, 95%CI 1.05-1.87 for the smallest DWS quintile vs other combined quintiles, p=0.022). CONCLUSIONS DWS was independently associated with an increased risk of incident HF in a community-based cohort of African Americans. DWS could be used as a qualitative estimator of LV myocardial stiffness.
Collapse
Affiliation(s)
- Daisuke Kamimura
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Takeki Suzuki
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Wanmei Wang
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael D Winniford
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Amil M Shah
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Carlos J Rodriguez
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Internal Medicine - Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kenneth R Butler
- Division of Geriatric Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Thomas H Mosley
- Division of Geriatric Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
12
|
Zuluaga M, Gregnanin G, Cencetti C, Di Meo C, Gueguen V, Letourneur D, Meddahi-Pellé A, Pavon-Djavid G, Matricardi P. PVA/Dextran hydrogel patches as delivery system of antioxidant astaxanthin: a cardiovascular approach. ACTA ACUST UNITED AC 2017; 13:015020. [PMID: 28875946 DOI: 10.1088/1748-605x/aa8a86] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
After myocardial infarction, the heart's mechanical properties and its intrinsic capability to recover are compromised. To improve this recovery, several groups have developed cardiac patches based on different biomaterials strategies. Here, we developed polyvinylalcohol/dextran (PVA/Dex) elastic hydrogel patches, obtained through the freeze thawing (FT) process, with the aim to deliver locally a potent natural antioxidant molecule, astaxanthin, and to assist the heart's response against the generated myofibril stress. Extensive rheological and dynamo-mechanical characterization of the effect of the PVA molecular weight, number of freeze-thawing cycles and Dex addition on the mechanical properties of the resulting hydrogels, were carried out. Hydrogel systems based on PVA 145 kDa and PVA 47 kDa blended with Dex 40 kDa, were chosen as the most promising candidates for this application. In order to improve astaxanthin solubility, an inclusion system using hydroxypropyl-β-cyclodextrin was prepared. This system was posteriorly loaded within the PVA/Dex hydrogels. PVA145/Dex 1FT and PVA47/Dex 3FT showed the best rheological and mechanical properties when compared to the other studied systems; environmental scanning electron microscope and confocal imaging evidenced a porous structure of the hydrogels allowing astaxanthin release. In vitro cellular behavior was analyzed after 24 h of contact with astaxanthin-loaded hydrogels. In vivo subcutaneous biocompatibility was performed in rats using PVA145/Dex 1FT, as the best compromise between mechanical support and astaxanthin delivery. Finally, ex vivo and in vivo experiments showed good mechanical and compatibility properties of this hydrogel. The obtained results showed that the studied materials have a potential to be used as myocardial patches to assist infarcted heart mechanical function and to reduce oxidative stress by the in situ release of astaxanthin.
Collapse
Affiliation(s)
- M Zuluaga
- INSERM, U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Paris 13 University, Sorbonne Paris Cite 99, Av. Jean-Baptiste Clément, F-93430 Villetaneuse, France. INSERM, U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, CHU X. Bichat, 46 rue H. Huchard, F-75018 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Yamamoto M, Ishizu T, Seo Y, Suto Y, Sai S, Xu D, Murakoshi N, Kimura T, Kawakami Y, Aonuma K. Teneligliptin Prevents Cardiomyocyte Hypertrophy, Fibrosis, and Development of Hypertensive Heart Failure in Dahl Salt-Sensitive Rats. J Card Fail 2017; 24:53-60. [PMID: 28888840 DOI: 10.1016/j.cardfail.2017.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/13/2017] [Accepted: 09/01/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND We investigated the effects of the dipeptidyl peptidase 4 inhibitor teneligliptin on cardiac function and hemodynamics during heart failure in hypertensive model rats. METHODS AND RESULTS Fifty-five male Dahl salt-sensitive rats were divided into 4 groups: control group (0.3% NaCl chow; n = 13), hypertension (HT) group (8% NaCl chow; n = 20), HT-early TNL group (8% NaCl chow and teneligliptin from 6 weeks; n = 10), and HT-late TNL group (8% NaCl chow and teneligliptin from 10 weeks; n = 12). Hemodynamic measurement and tissue analyses were performed at 18 weeks. In all of the HT groups, systolic blood pressures were similarly elevated (P = .66) and heart weights similarly increased (P = .36) with and without TNL administration. LV end-diastolic dimension was significantly enlarged only in the HT-early TNL group compared with the control group (P = .025). Histologic analysis showed less fibrosis (P = .008) and cardiomyocyte widths (P = .009) in the HT-early TNL group compared with the HT group. On hemodynamic analysis, only the HT group showed significant LV end-diastolic pressure elevation (P = .049) and lung congestion (P < .001) compared with the control group. CONCLUSIONS These results suggest that teneligliptin prevents concentric LV hypertrophy, fibrosis, and development of congestive heart failure in Dahl salt-sensitive rats. Teneligliptin may inhibit pressure-overload hypertrophic adaption and result in LV eccentric hypertrophy with reduced LV ejection fraction.
Collapse
Affiliation(s)
- Masayoshi Yamamoto
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomoko Ishizu
- Department of Clinical Laboratory Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| | - Yoshihiro Seo
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshimi Suto
- Department of Medical Sciences, University of Tsukuba, Ibaraki, Japan
| | - Seika Sai
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Dongzhu Xu
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Taizo Kimura
- Department of Clinical Laboratory Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yasushi Kawakami
- Department of Clinical Laboratory Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Cardiovascular Division, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
14
|
Ramadan S, Paul N, Naguib HE. Standardized static and dynamic evaluation of myocardial tissue properties. ACTA ACUST UNITED AC 2017; 12:025013. [PMID: 28065929 DOI: 10.1088/1748-605x/aa57a5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Quantifying the mechanical behaviors of soft biological tissues is of considerable research interest. However, validity and reproducibility between different researchers and apparatus is questionable. This study aims to quantify the mechanical properties of myocardium while investigating methodologies that can standardize biological tissue testing. MATERIALS AND METHODS Tensile testing was performed to obtain Young's modulus and a dynamic mechanical analysis (DMA) determined the viscoelastic properties. A frequency range of 0.5 Hz (30bpm) to 3.5 Hz (210bpm) was analyzed. For tensile testing three different preconditioning settings were tested: no load, 0.05 N preload, and a cyclic preload at 2.5% strain and 10 cycles. Samples were placed in saline and tested at 37 °C. Five ovine and five porcine hearts were tested. RESULTS AND DISCUSSION Cyclic loading results in the most consistent moduli values. The modulus of ovine/porcine tissue was mean = 0.05/.06 MPa, SD = 0.02/0.03 MPa. The storage/loss modulus varied from = 0.02/0.003 MPa at 0.5 Hz to 0.04/0.008 MPa at 3.5 Hz; Stiffness increases linearly from 400 to 800 N m-1 with a tan delta around 0.175. CONCLUSIONS Static analysis of the mechanical properties of myocardial tissue confirms that; preconditioning is necessary for reproducibility, and DMA provides a platform for reproducible testing of soft biological tissues.
Collapse
Affiliation(s)
- Sherif Ramadan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Rosebrugh Building (RS), 164 College Street, Room 407, Toronto, Ontario M5S 3G9, Canada
| | | | | |
Collapse
|
15
|
Michaelides M, Georgiadou S, Constantinides C. In vivo epicardial force and strain characterisation in normal and MLP-knockout murine hearts. Physiol Meas 2015; 36:1573-90. [PMID: 26057415 DOI: 10.1088/0967-3334/36/7/1573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The study's objective is to quantify in vivo epicardial force and strain in the normal and transgenic myocardium using microsensors.Male mice (n = 39), including C57BL/6 (n = 26), 129/Sv (n = 5), wild-type (WT) C57 × 129Sv (n = 5), and muscle LIM protein (MLP) knock-out (n = 3), were studied under 1.5% isoflurane anaesthesia. Microsurgery allowed the placement of two piezoelectric crystals at longitudinal epicardial loci at the basal, middle, and apical LV regions, and the independent (and/or concurrent) placement of a cantilever force sensor. The findings demonstrate longitudinal contractile and relaxation strains that ranged between 4.8-9.3% in the basal, middle, and apical regions of C57BL/6 mice, and in the mid-ventricular regions of 129/Sv, WT, and MLP mice. Measured forces ranged between 3.1-8.9 mN. The technique's feasibility is also demonstrated in normal mice following afterload, occlusion-reperfusion challenges.Furthermore, the total mid-ventricular forces developed in MLP mice were significantly reduced compared to the WT controls (5.9 ± 0.4 versus 8.9 ± 0.2 mN, p < 0.0001), possibly owing to the fibrotic and stiffer myocardium. No significant strain differences were noted between WT and MLP mice.The possibility of quantifying in vivo force and strain from the normal murine heart is demonstrated with a potential usefulness in the characterisation of transgenic and diseased mice, where regional myocardial function may be significantly altered.
Collapse
Affiliation(s)
- M Michaelides
- Department of Mechanical and Manufacturing Engineering, School of Engineering, University of Cyprus, Nicosia, Cyprus. Lecturer, Department of Sport and Exercise Science, UCLan Cyprus, University Avenue 12-14, Pyla 7080, Cyprus
| | | | | |
Collapse
|
16
|
Evaluation of Cardiovascular Risk Factors in the Wistar Audiogenic Rat (WAR) Strain. PLoS One 2015; 10:e0129574. [PMID: 26029918 PMCID: PMC4450865 DOI: 10.1371/journal.pone.0129574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 05/11/2015] [Indexed: 11/24/2022] Open
Abstract
Introduction Risk factors for life-threatening cardiovascular events were evaluated in an experimental model of epilepsy, the Wistar Audiogenic Rat (WAR) strain. Methods We used long-term ECG recordings in conscious, one year old, WAR and Wistar control counterparts to evaluate spontaneous arrhythmias and heart rate variability, a tool to assess autonomic cardiac control. Ventricular function was also evaluated using the pressure-volume conductance system in anesthetized rats. Results Basal RR interval (RRi) was similar between WAR and Wistar rats (188±5 vs 199±6 ms). RRi variability strongly suggests that WAR present an autonomic imbalance with sympathetic overactivity, which is an isolated risk factor for cardiovascular events. Anesthetized WAR showed lower arterial pressure (92±3 vs 115±5 mmHg) and exhibited indices of systolic dysfunction, such as higher ventricle end-diastolic pressure (9.2±0.6 vs 5.6±1 mmHg) and volume (137±9 vs 68±9 μL) as well as lower rate of increase in ventricular pressure (5266±602 vs 7320±538 mmHg.s-1). Indices of diastolic cardiac function, such as lower rate of decrease in ventricular pressure (-5014±780 vs -7766±998 mmHg.s-1) and a higher slope of the linear relationship between end-diastolic pressure and volume (0.078±0.011 vs 0.036±0.011 mmHg.μL), were also found in WAR as compared to Wistar control rats. Moreover, Wistar rats had 3 to 6 ventricular ectopic beats, whereas WAR showed 15 to 30 ectopic beats out of the 20,000 beats analyzed in each rat. Conclusions The autonomic imbalance observed previously at younger age is also present in aged WAR and, additionally, a cardiac dysfunction was also observed in the rats. These findings make this experimental model of epilepsy a valuable tool to study risk factors for cardiovascular events in epilepsy.
Collapse
|
17
|
S-nitroso human serum albumin attenuates pulmonary hypertension, improves right ventricular–arterial coupling, and reduces oxidative stress in a chronic right ventricle volume overload model. J Heart Lung Transplant 2015; 34:479-88. [DOI: 10.1016/j.healun.2014.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/23/2014] [Accepted: 09/19/2014] [Indexed: 11/18/2022] Open
|
18
|
Yu H, Tang M, Yu J, Zhou X, Zeng L, Zhang S. Chronic vagus nerve stimulation improves left ventricular function in a canine model of chronic mitral regurgitation. J Transl Med 2014; 12:302. [PMID: 25366939 PMCID: PMC4228179 DOI: 10.1186/s12967-014-0302-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 10/21/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Autonomic dysfunction, characterized by sympathetic activation and vagal withdrawal, contributes to the progression of heart failure (HF). We hypothesized that chronic vagus nerve stimulation (VNS) could prevent left ventricular (LV) remodeling and dysfunction in a canine HF model induced by chronic mitral regurgitation (MR). METHODS AND RESULTS After the MR inducing procedure, 12 survived canines were randomly divided into the control (n = 6) and the VNS (n = 6) groups. At month 2, a VNS stimulator system was implanted in all canines. From month 3 to month 6, VNS therapy was applied in the VNS group but not in the control group. At month 6, compared with the control group, the canines in VNS group had significantly higher cardiac output (2.3 ± 0.3 versus 2.9 ± 0.4 L/min, P < 0.05, LV forward stroke volume (20.1 ± 3.7 versus 24.8 ± 3.9 ml, P < 0.05), and end-systolic stiffness constant (2.2 ± 0.3 versus 2.7 ± 0.3, P < 0.05). NT-proBNP and C-reactive protein were decreased significantly in the VNS group. However, no statistical difference was found in LV ejection fraction, LV end-diastolic dimension, LV end-diastolic volume, myocyte cross-sectional area, or collagen volume fraction between two groups. CONCLUSIONS Chronic VNS therapy may ameliorate MR-induced LV contractile dysfunction and improve the expression of biomarkers, but has less effect in improving LV chamber remodeling.
Collapse
Affiliation(s)
- Haiwen Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100037, People's Republic of China.
| | - Min Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100037, People's Republic of China.
| | - Jun Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100037, People's Republic of China.
| | - Xiaohong Zhou
- Cardiac Rhythm Disease Management, Chinese Branch of Medtronic Inc., Shanghai, China.
| | - Lepeng Zeng
- Cardiac Rhythm Disease Management, Chinese Branch of Medtronic Inc., Shanghai, China.
| | - Shu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100037, People's Republic of China.
| |
Collapse
|
19
|
Kanis LA, Marques EL, Zepon KM, Pereira JR, Pamato S, de Oliveira MT, Danielski LG, Petronilho FC. Cellulose acetate butyrate/poly(caprolactonetriol) blends: Miscibility, mechanical properties, and in vivo inflammatory response. J Biomater Appl 2014; 29:654-61. [PMID: 25015652 DOI: 10.1177/0885328214542488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study reports the results of the characterization of cellulose acetate butyrate and polycaprolactone-triol blends in terms of miscibility, swelling capacity, mechanical properties, and inflammatory response in vivo. The cellulose acetate butyrate film was opaque and rigid, with glass transition (T g ) at 134℃ and melting temperature of 156℃. The cellulose acetate butyrate/polycaprolactone-triol films were transparent up to a polycaprolactone-triol content of 60%. T g of the cellulose acetate butyrate films decreased monotonically as polycaprolactone-triol was added to the blend, thus indicating miscibility. FTIR spectroscopy revealed a decrease in intramolecular hydrogen bonding in polycaprolactone-triol, whereas no hydrogen bonding was observed between cellulose acetate butyrate and -OH from polycaprolactone-triol. The increase in polycaprolactone-triol content in the blend decreased the water uptake. An increase in polycaprolactone-triol content decreased the modulus of elasticity and increased the elongation at break. A cellulose acetate butyrate/polycaprolactone-triol 70/30 blend implanted in rats showed only an acute inflammatory response 7 days after surgery. No change in inflammation mediators was observed.
Collapse
Affiliation(s)
- Luiz A Kanis
- TECFARMA, University of Southern Santa Catarina, Tubarão, SC, Brazil Post Graduated Program in Health Science, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Ellen L Marques
- TECFARMA, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Karine M Zepon
- TECFARMA, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Jefferson R Pereira
- Post Graduated Program in Health Science, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Saulo Pamato
- Post Graduated Program in Health Science, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Marcelo T de Oliveira
- Post Graduated Program in Health Science, University of Southern Santa Catarina, Tubarão, SC, Brazil
| | - Lucinéia G Danielski
- LAFICEXP, Universidade University of Southern Santa Catarina, Tubarão, SC, Brazil
| | | |
Collapse
|
20
|
Chronic overcirculation-induced pulmonary arterial hypertension in aorto-caval shunt. Microvasc Res 2014; 94:73-9. [PMID: 24862700 DOI: 10.1016/j.mvr.2014.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 05/09/2014] [Accepted: 05/15/2014] [Indexed: 11/20/2022]
Abstract
Pulmonary arterial hypertension is a common complication of congenital heart defects with left-to-right shunts. Current preclinical models do not reproduce clinical characteristics of shunt-related pulmonary hypertension. Aorto-caval shunt was firstly described as a model of right ventricle volume overload. The pathophysiology and the possible determination of pulmonary arterial hypertension of different periods of shunt exposure are still undefined. A method to create standardized, reproducible aorto-caval shunt was developed in growing rats (260±40 g). Three groups of animals were considered: shunt exposure for 10 weeks, shunt exposure for 20 weeks and control (sham laparotomy). Echocardiography and magnetic resonance revealed increased right ventricular end diastolic area in shunt at 10 weeks compared to control. Hemodynamic analysis demonstrated increased right ventricular afterload and increased effective pulmonary arterial elastance (Ea) in shunt at 20 weeks compared to control (1.29±0.20 vs. 0.14±0.06 mmHg/μl, p=0.004). At the same time point, the maximal slope of end-systolic pressure-volume relationship (Ees) decreased (0.5±0.2 mmHg/ml vs. 1.2±0.3, p<0.001). Consequently, right ventricular-arterial coupling was markedly deteriorated with a ≈50% decrease in the ratio of end-systolic to pulmonary artery elastance (Ees/Ea). Finally, left ventricular preload diminished (≈30% decrease in left ventricular end-diastolic volume). Histology demonstrated medial hypertrophy and small artery luminal narrowing. Chronic exposure to aorto-caval shunt is a reliable model to produce right ventricular volume overload and secondary pulmonary arterial hypertension. This model could be an alternative with low mortality and high reproducibility for investigators on the underlying mechanisms of shunt-related pulmonary hypertension.
Collapse
|
21
|
Ventricular and pulmonary vascular remodeling induced by pulmonary overflow in a chronic model of pretricuspid shunt. J Thorac Cardiovasc Surg 2014; 148:2609-17. [PMID: 24908349 DOI: 10.1016/j.jtcvs.2014.04.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 04/14/2014] [Accepted: 04/24/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Current preclinical models of pulmonary arterial hypertension do not reproduce the clinical characteristics of congenital heart anomalies. Aortocaval shunt is relevant to a variety of clinical conditions. The pathophysiology and possible determination of pulmonary hypertension in this model are still undefined. METHODS A method to create a standardized and reproducible aortocaval shunt was developed in rats. After creation of the shunt, the animals were followed up for 20 weeks and a sham laparotomy was used as a control. The chronic effects of volume overload on the right and left ventricles and pulmonary hemodynamic modifications were evaluated by biventricular catheterization, echocardiography, and magnetic resonance. Pulmonary vascular changes were defined by histology. RESULTS An increased right ventricular end-diastolic area was confirmed by echocardiography. Left ventricular overload and decreased biventricular ejection fraction were demonstrated by magnetic resonance after 20 weeks in the shunt group compared with the controls (left ventricle, 50% ± 5% vs 62% ± 3%, P = .029; right ventricle, 53% ± 2% vs 65% ± 2%, P = .036). Preload recruitable stroke work of left and right ventricles decreased after 20 weeks in shunt rats (left ventricle: 36 ± 7 vs 98 ± 5, P = .004; right ventricle: 19 ± 2 vs 32 ± 9, P = .047). At the same time point, catheterization showed that effective pulmonary arterial elastance was increased only in the shunt group (1.29 ± 0.20 vs 0.14 ± 0.06 mm Hg/μL; P = .004). Histology showed medial hypertrophy, small artery luminal narrowing, and occlusion. CONCLUSIONS The aortocaval shunt model reliably produces right ventricular volume overload and secondary pulmonary hypertension. Due to a combination of left ventricular dysfunction and pulmonary overflow, the pulmonary hypertension produced shows features similar to those found in patients with chronic atrial-level shunt.
Collapse
|
22
|
Ghasemi-Mobarakeh L, Prabhakaran MP, Nematollahi M, Karbalaie K, Ramakrishna S, Nasr-Esfahani MH. Embryonic Stem Cell Differentiation to Cardiomyocytes on Nanostructured Scaffolds for Myocardial Tissue Regeneration. INT J POLYM MATER PO 2013. [DOI: 10.1080/00914037.2013.830247] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Jia X, Choy JS, Zhang ZD, Svendsen M, Zhong L, Tan RS, Kassab GS. Extent of load-independence of pressure-normalized stress in swine. Exp Biol Med (Maywood) 2013; 238:821-9. [PMID: 23828596 DOI: 10.1177/1535370213494548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A load-independent index of myocardial contractility provides a measure of cardiac function. Previous contractility indices have been shown to be either load-dependent or invasive. We sought to determine the extent of load (preload and afterload)-independence of dσ*/dtmax (σ* is pressure-normalized stress) in comparison with other well-established indices. Six anaesthetized pigs underwent left ventricular pressure-volume measurements under various load conditions. The average preload was decreased by 70.0 ± 15.0% (from 39.2 ± 6.4 mL to 11.7 ± 7.7 mL) and increased by 49.3 ± 5.9% (from 35.1 ± 7.4 mL to 51.7 ± 8.9 mL). The average afterload was increased by 74.3 ± 43.5% (from 3.3 ± 0.6 mmHg/mL to 5.7 ± 1.7 mmHg/mL). When preload was reduced within an average of 21.7% (39.2 ± 6.4 mL to 30.7 ± 6.2 mL) using occlusion of the inferior vena cava, dσ*/dt max did not change significantly (6.50 ± 1.10 s⁻¹ vs 6.60 ± 0.90 s⁻¹, P = non-significant [NS]). When preload was increased within an average of 29.3% (35.1 ± 7.4 mL to 45.4 ± 7.3 mL) from infusion of normal saline, dσ*/dt max did not change significantly (7.04 ± 1.00 s⁻¹ vs 7.29 ± 1.10 s⁻¹, P = NS). When afterload was increased within an average of 42.4% (3.3 ± 0.6 mmHg/mL to 4.7 ± 1.0 mmHg/mL) using intra-aortic balloon occlusion, dσ*/dtmax did not change significantly (6.72 ± 1.18 s⁻¹ vs 6.89 ± 1.28 s⁻¹, P = NS). As expected, dσ*/dtmax was significantly increased with dobutamine. A linear regression showed no correlation between dσ*/dtmax and preload (r² = 0.02, P = 0.17) within a maximum range of -30% to +50% of preload change, or between dσ*/dtmax and afterload (r² = 0.03, P = 0.36) within maximum range of 0-100% of afterload increase, respectively. In conclusion, dσ*/dtmax is independent of loading conditions within an average of 21.7% of preload decrease, 29.3% of preload increase, 42.4% of afterload increase, and sensitive to dobutamine infusion.
Collapse
Affiliation(s)
- Xinwei Jia
- Department of Biomedical Engineering, Indiana University - Purdue University Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Venugopal J, Rajeswari R, Shayanti M, Sridhar R, Sundarrajan S, Balamurugan R, Ramakrishna S. Xylan polysaccharides fabricated into nanofibrous substrate for myocardial infarction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:1325-31. [DOI: 10.1016/j.msec.2012.12.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 09/21/2012] [Accepted: 12/04/2012] [Indexed: 11/30/2022]
|
25
|
|
26
|
Patel A, Gaharwar AK, Iviglia G, Zhang H, Mukundan S, Mihaila SM, Demarchi D, Khademhosseini A. Highly elastomeric poly(glycerol sebacate)-co-poly(ethylene glycol) amphiphilic block copolymers. Biomaterials 2013; 34:3970-3983. [PMID: 23453201 DOI: 10.1016/j.biomaterials.2013.01.045] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/07/2013] [Indexed: 12/13/2022]
Abstract
Poly(glycerol sebacate) (PGS), a tough elastomer, has been proposed for tissue engineering applications due to its desired mechanical properties, biocompatibility and controlled degradation. Despite interesting physical and chemical properties, PGS shows limited water uptake capacity (∼2%), thus constraining its utility for soft tissue engineering. Therefore, a modification of PGS that would mimic the water uptake and water retention characteristics of natural extracellular matrix is beneficial for enhancing its utility for biomedical applications. Here, we report the synthesis and characterization of highly elastomeric poly(glycerol sebacate)-co-polyethylene glycol (PGS-co-PEG) block copolymers with controlled water uptake characteristics. By tailoring the water uptake property, it is possible to engineer scaffolds with customized degradation and mechanical properties. The addition of PEG results in almost 15-fold increase in water uptake capacity of PGS, and improves its mechanical stability under dynamic loading conditions. PGS-co-PEG polymers show elastomeric properties and can be subjected to serve deformation such as bending and stretching. The Young's modulus of PGS-co-PEG can be tuned from 13 kPa to 2.2 MPa by altering the amount of PEG within the copolymer network. Compared to PGS, more than six-fold increase in elongation was observed upon PEG incorporation. In addition, the rate of degradation increases with an increase in PEG concentration, indicating that degradation rate of PGS can be regulated. PGS-co-PEG polymers also support cell proliferation, and thus can be used for a range of tissue engineering applications.
Collapse
Affiliation(s)
- Alpesh Patel
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Akhilesh K Gaharwar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giorgio Iviglia
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electronics and Telecommunications, Politecnico di Torino, Torino 10129, Italy
| | - Hongbin Zhang
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; School of Materials Science and Engineering, University of Science and Technology, Beijing, Beijing 100083, China
| | - Shilpaa Mukundan
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Silvia M Mihaila
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, Torino 10129, Italy; Center for Space Human Robotics, Italian Institute of Technology, Torino, Italy
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
27
|
Myocardial contractile dysfunction associated with increased 3-month and 1-year mortality in hospitalized patients with heart failure and preserved ejection fraction. Int J Cardiol 2013; 168:1975-83. [PMID: 23336957 DOI: 10.1016/j.ijcard.2012.12.084] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 09/12/2012] [Accepted: 12/27/2012] [Indexed: 11/21/2022]
Abstract
BACKGROUND There is a clinical need for a contractility index that reflects myocardial contractile dysfunction even when ejection fraction (EF) is preserved. We used novel relative load-independent global and regional contractility indices to compare left ventricular (LV) contractile function in three groups: heart failure (HF) with preserved ejection fraction (HFPEF), HF with reduced ejection fraction (HFREF) and normal subjects. Also, we determined the associations of these parameters with 3-month and 1-year mortality in HFPEF patients. METHODS 199 HFPEF patients [median age (IQR): 75 (67-80) years] and 327 HFREF patients [69 (59-76) years] were recruited following hospitalization for HF; 22 normal control subjects [65 (54-71) years] were recruited for comparison. All patients underwent standard two-dimensional Doppler and tissue Doppler echocardiography to characterize LV dimension, structure, global and regional contractile function. RESULTS The median (IQR) global LV contractility index, dσ*/dtmax was 4.30s(-1) (3.51-4.57s(-1)) in normal subjects but reduced in HFPEF [2.57 (2.08-3.64)] and HFREF patients [1.77 (1.34-2.30)]. Similarly, median (IQR) regional LV contractility index was 99% (88-104%) in normal subjects and reduced in HFPEF [81% (66-96%)] and HFREF [56% (41-71%)] patients. Multi-variable logistic regression analysis on HFPEF identified sc-mFS <76% as the most consistent predictor of both 3-month (OR=7.15, p<0.05) and 1-year (OR=2.57, p<0.05) mortality after adjusting for medical conditions and other echocardiographic measurements. CONCLUSION Patients with HFPEF exhibited decreased LV global and regional contractility. This population-based study demonstrated that depressed regional contractility index was associated with higher 3-month and 1-year mortality in HFPEF patients.
Collapse
|
28
|
Abstract
Interactions between the left ventricle (LV) and the arterial system, (ventricular-arterial coupling) are key determinants of cardiovascular function. Ventricularearterial coupling is most frequently assessed in the pressure-volume plane using the ratio of effective arterial elastance (EA) to LV end-systolic elastance (EES). EA (usually interpreted as a lumped index of arterial load) can be computed as end-systolic pressure/stroke volume, whereas EES (a load-independent measure of LV chamber systolic stiffness and contractility) is ideally assessed invasively using data from a family of pressure-volume loops obtained during an acute preload alteration. Single-beat methods have also been proposed, allowing for non-invasive estimations of EES using simple echocardiographic measurements. The EA/EES ratio is useful because it provides information regarding the operating mechanical efficiency and performance of the ventricular-arterial system. However, it should be recognized that analyses in the pressure-volume plane have several limitations and that "ventricular-arterial coupling" encompasses multiple physiologic aspects, many of which are not captured in the pressure-volume plane. Therefore, additional assessments provide important incremental physiologic information about the cardiovascular system and should be more widely used. In particular, it should be recognized that: (1) comprehensive analyses of arterial load are important because EA poorly characterizes pulsatile LV load and does not depend exclusively on arterial properties; (2) The systolic loading sequence, an important aspect of ventricular-arterial coupling, is neglected by pressure-volume analyses, and can profoundly impact LV function, remodeling and progression to heart failure. This brief review summarizes methods for the assessment of ventricular-arterial interactions, as discussed at the Artery 12 meeting (October 2012).
Collapse
Affiliation(s)
- Julio A Chirinos
- University of Pennsylvania, Philadelphia, PA, USA ; Philadelphia VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
29
|
Ravichandran R, Venugopal JR, Sundarrajan S, Mukherjee S, Sridhar R, Ramakrishna S. Minimally invasive injectable short nanofibers of poly(glycerol sebacate) for cardiac tissue engineering. NANOTECHNOLOGY 2012; 23:385102. [PMID: 22947662 DOI: 10.1088/0957-4484/23/38/385102] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Myocardial tissue lacks the ability to appreciably regenerate itself following myocardial infarction (MI) which ultimately results in heart failure. Current therapies can only retard the progression of disease and hence tissue engineering strategies are required to facilitate the engineering of a suitable biomaterial to repair MI. The aim of this study was to investigate the in vitro properties of an injectable biomaterial for the regeneration of infarcted myocardium. Fabrication of core/shell fibers was by co-axial electrospinning, with poly(glycerol sebacate) (PGS) as core material and poly-L-lactic acid (PLLA) as shell material. The PLLA was removed by treatment of the PGS/PLLA core/shell fibers with DCM:hexane (2:1) to obtain PGS short fibers. These PGS short fibers offer the advantage of providing a minimally invasive injectable technique for the regeneration of infarcted myocardium. The scaffolds were characterized by SEM, FTIR and contact angle and cell-scaffold interactions using cardiomyocytes. The results showed that the cardiac marker proteins actinin, troponin, myosin heavy chain and connexin 43 were expressed more on short PGS fibers compared to PLLA nanofibers. We hypothesized that the injection of cells along with short PGS fibers would increase cell transplant retention and survival within the infarct, compared to the standard cell injection system.
Collapse
Affiliation(s)
- Rajeswari Ravichandran
- Healthcare and Energy Materials Laboratory, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
30
|
Ishii R, Higashimori M, Tadakuma K, Kaneko M, Tamaki S, Sakata Y, Yamamoto K. Balloon type elasticity sensing for left ventricle of small laboratory animal. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2011:904-7. [PMID: 22254457 DOI: 10.1109/iembs.2011.6090202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This paper describes an elasticity sensing system for left ventricle of small laboratory animal. We first show the basic concept of the proposed method, where a ring shaped specimen is dilated by a balloon type probe using a pressure based control, and the elasticity of the specimen is estimated by using the stress and strain information. We introduce a dual cylinder model for approximating the strengths of the specimen's material and the balloon. Based on this model, we can derive Young's modulus of the specimen. After explaining the developed experimental system, we show a couple of experimental results using rats and mice, where HFPEF (Heart Failure Preserved Ejection Fraction) group can be distinguished from a normal group.
Collapse
Affiliation(s)
- Ryohei Ishii
- Department of Mechanical Engineering, Osaka University, 2-1 Yamadaoka, Suita, Japan. ishii@hh
| | | | | | | | | | | | | |
Collapse
|
31
|
Bozkurt B, Bolos M, Deswal A, Ather S, Chan W, Mann DL, Carabello B. New Insights into Mechanisms of Action of Carvedilol Treatment in Chronic Heart Failure Patients—A Matter of Time for Contractility. J Card Fail 2012; 18:183-93. [DOI: 10.1016/j.cardfail.2011.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 11/26/2011] [Accepted: 11/30/2011] [Indexed: 10/14/2022]
|
32
|
GHISTA DHANJOON, ZHONG LIANG, LE THUTHAO, TAN RUSAN. CARDIAC CONTRACTILITY MEASURES OF LEFT VENTRICULAR SYSTOLIC FUNCTIONAL ASSESSMENT OF NORMAL AND DISEASED HEARTS. J MECH MED BIOL 2011. [DOI: 10.1142/s0219519409003139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Left ventricular (LV) contraction is the basis of LV systolic function, impairment of which underlies heart failure pathophysiology. Its accurate quantification in the form of LV contractility indices is imperative for diagnostic and follow-up assessment of LV systolic function in heart failure. Herein, we analyze LV contractile performance by focusing on LV contractility indices at different physiological organizational levels: from sarcomere dynamics to LV myocardial properties (such as elastic modulus and elastance), and from LV wall contractile stress development to the generation of intra-LV blood flow velocities and pressure distributions. Further, we present the development analyses of these indices and their medical applications. Using improved development of invasive and noninvasive techniques for measuring ventricular pressure, geometry, and volume, we show how these indices have become more amenable for clinical usage to obtain better patient assessment. The purpose of this paper is to present a comprehensive coverage of LV contraction physiology, indices to qualify LV contraction, formulation, and medical applications of some major intrinsic LV contractility indices, so as to provide the basis of functional assessment of normal versus diseased hearts.
Collapse
Affiliation(s)
| | - LIANG ZHONG
- Department of Cardiology, National Heart Centre, Mistri Wing, 17 Third Hospital Avenue, Singapore
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - THU-THAO LE
- Research and Development Unit, National Heart Centre, Mistri Wing, 17 Third Hospital Avenue, Singapore
| | - RU-SAN TAN
- Department of Cardiology, National Heart Centre, Mistri Wing, 17 Third Hospital Avenue, Singapore
| |
Collapse
|
33
|
Venugopal JR, Prabhakaran MP, Mukherjee S, Ravichandran R, Dan K, Ramakrishna S. Biomaterial strategies for alleviation of myocardial infarction. J R Soc Interface 2011; 9:1-19. [PMID: 21900319 PMCID: PMC3223634 DOI: 10.1098/rsif.2011.0301] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
World Health Organization estimated that heart failure initiated by coronary artery disease and myocardial infarction (MI) leads to 29 per cent of deaths worldwide. Heart failure is one of the leading causes of death in industrialized countries and is expected to become a global epidemic within the twenty-first century. MI, the main cause of heart failure, leads to a loss of cardiac tissue impairment of left ventricular function. The damaged left ventricle undergoes progressive ‘remodelling’ and chamber dilation, with myocyte slippage and fibroblast proliferation. Repair of diseased myocardium with in vitro-engineered cardiac muscle patch/injectable biopolymers with cells may become a viable option for heart failure patients. These events reflect an apparent lack of effective intrinsic mechanism for myocardial repair and regeneration. Motivated by the desire to develop minimally invasive procedures, the last 10 years observed growing efforts to develop injectable biomaterials with and without cells to treat cardiac failure. Biomaterials evaluated include alginate, fibrin, collagen, chitosan, self-assembling peptides, biopolymers and a range of synthetic hydrogels. The ultimate goal in therapeutic cardiac tissue engineering is to generate biocompatible, non-immunogenic heart muscle with morphological and functional properties similar to natural myocardium to repair MI. This review summarizes the properties of biomaterial substrates having sufficient mechanical stability, which stimulates the native collagen fibril structure for differentiating pluripotent stem cells and mesenchymal stem cells into cardiomyocytes for cardiac tissue engineering.
Collapse
Affiliation(s)
- Jayarama Reddy Venugopal
- Healthcare and Energy Materials Laboratory, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
34
|
Rai R, Boccaccini AR, Knowles JC, Mordon N, Salih V, Locke IC, Moshrefi-Torbati M, Keshavarz T, Roy I. The homopolymer poly(3-hydroxyoctanoate) as a matrix material for soft tissue engineering. J Appl Polym Sci 2011. [DOI: 10.1002/app.34772] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Zhong L, Poh KK, Lee LC, Le TT, Tan RS. Attenuation of Stress-based Ventricular Contractility in Patients with Heart Failure and Normal Ejection Fraction. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2011. [DOI: 10.47102/annals-acadmedsg.v40n4p179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: The maximal rate of change of pressure-normalised wall stress dσ*/dtmax has been proposed as cardiac index of left ventricular (LV) contractility. In this study, we assessed the capacity of dσ*/dtmax to diagnose heart failure with normal ejection fraction (HFNEF). Materials and Methods: One hundred healthy normal controls and 140 patients admitted with heart failure (100, HFREF and 40, HFNEF) underwent echocardiography for stress-based contractility dσ*/dtmax. Patients with significant valvular heart disease were excluded. Tissue Doppler indices were also measured. Results: dσ*/dtmaxwas 4.43 ± 1.27 s-1 in control subjects; reduced in HFNEF, 3.02 ± 0.98 s-1; and HFREF, 2.00 ± 0.67 s-1 (P <0.001). In comparison with age- and sex-matched groups (n = 26 each), we found similar trend on reduction of dσ*/dtmax (normal control; 3.91 ± 0.87 s-1; HFNEF, 2.90 ± 0.84 s-1; HFREF, 1.84 ± 0.59 s-1, P <0.001). On multivariate analysis, dσ*/dtmax was found to be the independent predictor of HFNEF and HFREF. The area under the curve of the receiver operating characteristics (ROC) in detecting HFNEF compared with normal controls (dσ*/dtmax >3.2 s-1) was 0.84 (P <0.0001), and in detecting HFREF compared with HFNEF (dσ*/dtmax >2.32 s-1) was 0.88 (P <0.0001). Conclusion: This data confirms that dσ*/dtmax on echocardiography is a powerful independent predictor in patients with HFNEF. In a population with a high suspicion of HFNEF, dσ*/dtmax may significantly contribute to early diagnosis and hence be useful in the triage and management of HFNEF patients.
Key words: Echocardiography, Heart failure with normal ejection fraction, Normalised wall-stress
Collapse
|
36
|
Ou L, Li W, Zhang Y, Wang W, Liu J, Sorg H, Furlani D, Gäbel R, Mark P, Klopsch C, Wang L, Lützow K, Lendlein A, Wagner K, Klee D, Liebold A, Li RK, Kong D, Steinhoff G, Ma N. Intracardiac injection of matrigel induces stem cell recruitment and improves cardiac functions in a rat myocardial infarction model. J Cell Mol Med 2010; 15:1310-8. [PMID: 20477905 PMCID: PMC4373331 DOI: 10.1111/j.1582-4934.2010.01086.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Matrigel promotes angiogenesis in the myocardium from ischemic injury and prevents remodelling of the left ventricle. We assessed the therapeutic efficacy of intracardiac matrigel injection and matrigel-mediated stem cell homing in a rat myocardial infarction (MI) model. Following MI, matrigel (250 μl) or phosphate-buffered solution (PBS) was delivered by intracardiac injection. Compared to the MI control group (MI-PBS), matrigel significantly improved left ventricular function (n= 11, P < 0.05) assessed by pressure–volume loops after 4 weeks. There is no significant difference in infarct size between MI-matrigel (MI-M; 21.48 ± 1.49%, n= 10) and MI-PBS hearts (20.98 ± 1.25%, n= 10). The infarct wall thickness of left ventricle is significantly higher (P < 0.01) in MI-M (0.72 ± 0.02 mm, n= 10) compared with MI-PBS (0.62 ± 0.02 mm, n= 10). MI-M hearts exhibited higher capillary density (border 130.8 ± 4.7 versus 115.4 ± 6.0, P < 0.05; vessels per high-power field [HPF; 400×], n= 6) than MI-PBS hearts. c-Kit+ stem cells (38.3 ± 5.3 versus 25.7 ± 1.5 c-Kit+ cells per HPF [630×], n= 5, P < 0.05) and CD34+ cells (13.0 ± 1.51 versus 5.6 ± 0.68 CD34+ cells per HPF [630×], n= 5, P < 0.01) were significantly more numerous in MI-M than in MI-PBS in the infarcted hearts (n= 5, P < 0.05). Intracardiac matrigel injection restores myocardial functions following MI, which may attribute to the improved recruitment of CD34+ and c-Kit+ stem cells.
Collapse
Affiliation(s)
- Lailiang Ou
- Reference- and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fayssoil A, Renault G, Fougerousse F. End-systolic stress–velocity relation and circumferential fiber velocity shortening for analysing left ventricular function in mice. Radiography (Lond) 2009. [DOI: 10.1016/j.radi.2008.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
38
|
Mukhopadhyay P, Rajesh M, Bátkai S, Kashiwaya Y, Haskó G, Liaudet L, Szabó C, Pacher P. Role of superoxide, nitric oxide, and peroxynitrite in doxorubicin-induced cell death in vivo and in vitro. Am J Physiol Heart Circ Physiol 2009; 296:H1466-83. [PMID: 19286953 PMCID: PMC2685360 DOI: 10.1152/ajpheart.00795.2008] [Citation(s) in RCA: 284] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 03/09/2009] [Indexed: 01/11/2023]
Abstract
Doxorubicin (DOX) is a potent available antitumor agent; however, its clinical use is limited because of its cardiotoxicity. Cell death is a key component in DOX-induced cardiotoxicity, but its mechanisms are elusive. Here, we explore the role of superoxide, nitric oxide (NO), and peroxynitrite in DOX-induced cell death using both in vivo and in vitro models of cardiotoxicity. Western blot analysis, real-time PCR, immunohistochemistry, flow cytometry, fluorescent microscopy, and biochemical assays were used to determine the markers of apoptosis/necrosis and sources of NO and superoxide and their production. Left ventricular function was measured by a pressure-volume system. We demonstrated increases in myocardial apoptosis (caspase-3 cleavage/activity, cytochrome c release, and TUNEL), inducible NO synthase (iNOS) expression, mitochondrial superoxide generation, 3-nitrotyrosine (NT) formation, matrix metalloproteinase (MMP)-2/MMP-9 gene expression, poly(ADP-ribose) polymerase activation [without major changes in NAD(P)H oxidase isoform 1, NAD(P)H oxidase isoform 2, p22(phox), p40(phox), p47(phox), p67(phox), xanthine oxidase, endothelial NOS, and neuronal NOS expression] and decreases in myocardial contractility, catalase, and glutathione peroxidase activities 5 days after DOX treatment to mice. All these effects of DOX were markedly attenuated by peroxynitrite scavengers. Doxorubicin dose dependently increased mitochondrial superoxide and NT generation and apoptosis/necrosis in cardiac-derived H9c2 cells. DOX- or peroxynitrite-induced apoptosis/necrosis positively correlated with intracellular NT formation and could be abolished by peroxynitrite scavengers. DOX-induced cell death and NT formation were also attenuated by selective iNOS inhibitors or in iNOS knockout mice. Various NO donors when coadministered with DOX but not alone dramatically enhanced DOX-induced cell death with concomitant increased NT formation. DOX-induced cell death was also attenuated by cell-permeable SOD but not by cell-permeable catalase, the xanthine oxidase inhibitor allopurinol, or the NADPH oxidase inhibitors apocynine or diphenylene iodonium. Thus, peroxynitrite is a major trigger of DOX-induced cell death both in vivo and in vivo, and the modulation of the pathways leading to its generation or its effective neutralization can be of significant therapeutic benefit.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/toxicity
- Antioxidants/metabolism
- Apoptosis/drug effects
- Cell Line
- Dose-Response Relationship, Drug
- Doxorubicin/toxicity
- Enzyme Inhibitors/pharmacology
- Free Radical Scavengers/pharmacology
- Heart Diseases/chemically induced
- Heart Diseases/metabolism
- Heart Diseases/pathology
- Heart Diseases/physiopathology
- Heart Diseases/prevention & control
- Male
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Necrosis
- Nitric Oxide/metabolism
- Nitric Oxide Donors/pharmacology
- Nitric Oxide Synthase Type II/antagonists & inhibitors
- Nitric Oxide Synthase Type II/deficiency
- Nitric Oxide Synthase Type II/genetics
- Peroxynitrous Acid/metabolism
- Poly(ADP-ribose) Polymerases/metabolism
- Superoxides/metabolism
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- Ventricular Function, Left/drug effects
- Ventricular Pressure/drug effects
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Section on Oxidative Stress and Tissue Injury, Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, MSC-9413, Bethesda, MD 20892-9413, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Effects of surgical ventricular restoration on left ventricular contractility assessed by a novel contractility index in patients with ischemic cardiomyopathy. Am J Cardiol 2009; 103:674-9. [PMID: 19231332 DOI: 10.1016/j.amjcard.2008.10.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 10/31/2008] [Accepted: 10/31/2008] [Indexed: 11/22/2022]
Abstract
A pressure-normalized left ventricular (LV) wall stress (dsigma*/dt(max)) was recently reported as a load-independent index of LV contractility. We hypothesized that this novel contractility index might demonstrate improvement in LV contractile function after surgical ventricular restoration (SVR) using magnetic resonance imaging. A retrospective analysis of magnetic resonance imaging data of 40 patients with ischemic cardiomyopathy who had undergone coronary artery bypass grafting with SVR was performed. LV volumes, ejection fraction, global systolic and diastolic sphericity, and dsigma*/dt(max) were calculated. After SVR, a decrease was found in end-diastolic and end-systolic volume indexes, whereas LV ejection fraction increased from 26% +/- 7% to 31% +/- 10% (p <0.001). LV mass index and peak normalized wall stress were decreased, whereas the sphericity index (SI) at end-diastole increased, indicating that the left ventricle became more spherical after SVR. LV contractility index dsigma*/dt(max) improvement (from 2.69 +/- 0.74 to 3.23 +/- 0.73 s(-1), p <0.001) was associated with shape change as evaluated by the difference in SI between diastole and systole (r = 0.32, p <0.001, preoperative; r = 0.23, p <0.001, postoperative), but not with baseline LV SI. In conclusion, SVR excludes akinetic LV segments and decreases LV wall stress. Despite an increase in sphericity, LV contractility, as determined by dsigma*/dt(max), actually improves. A complex interaction of LV maximal flow rate and LV mass may explain the improvement in LV contractility after SVR. Because dsigma*/dt(max) can be estimated from simple noninvasive measurements, this underscores its clinical utility for assessment of contractile function with therapeutic intervention.
Collapse
|
40
|
Transition of ventricular function and energy efficiency after a primary or staged Fontan procedure. Gen Thorac Cardiovasc Surg 2008; 56:498-504. [PMID: 18854926 DOI: 10.1007/s11748-008-0292-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 06/18/2008] [Indexed: 10/21/2022]
Abstract
PURPOSE The transitional changes of the ventricular function at different time points after total cavopulmonary connection (TCPC) were examined. METHODS A total of 29 patients were divided into a primary TCPC group and a staged TCPC group. In both groups, phase I was defined as within 2 months after TCPC and phase II as beyond at least a year after TCPC. Changes in ventricular end-diastolic volume (EDV), ventricular end-systolic elastance (Ees), effective arterial elastance (Ea), and ventriculoarterial coupling (Ea/Ees) were evaluated. RESULTS The results for the primary TCPC group are as follows. Phase I: The EDV decreased (P < 0.05). The Ees and Ea both increased (P < 0.05). Ea/Ees showed a tendency to increase (P = 0.08). Phase II: The EDV decreased (P < 0.05). The Ees increased significantly, and Ea showed no significant change. Ees/Ea showed a tendency to decrease (P = 0.07). The results for the staged TCPC group were as follows. Phase I: The EDV decreased significantly after bidirectional cavopulmonary shunt (BCPS). The Ees showed no significant change after BCPS and TCPC. Although Ea increased after BCPS (P < 0.05), it showed no significant change after TCPC. Ea/Ees showed no significant change. Phase II: The Ees increased (P < 0.05) without significant changes of EDV and Ea. As a result, Ea/Ees showed a tendency to decrease. CONCLUSION This study suggested improved ventricular function in both groups. These results suggest hemodynamic adaptation to the Fontan circulation. The deleterious effects on ventricular function caused by the Fontan procedure disappeared within a couple of years. This acute effect can be ameliorated by the staged approach to the Fontan circulation.
Collapse
|
41
|
Rosner A, Bijnens B, Hansen M, How OJ, Aarsaether E, Muller S, Sutherland GR, Myrmel T. Left ventricular size determines tissue Doppler-derived longitudinal strain and strain rate. EUROPEAN JOURNAL OF ECHOCARDIOGRAPHY 2008; 10:271-7. [DOI: 10.1093/ejechocard/jen230] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
42
|
Chen QZ, Bismarck A, Hansen U, Junaid S, Tran MQ, Harding SE, Ali NN, Boccaccini AR. Characterisation of a soft elastomer poly(glycerol sebacate) designed to match the mechanical properties of myocardial tissue. Biomaterials 2008; 29:47-57. [PMID: 17915309 DOI: 10.1016/j.biomaterials.2007.09.010] [Citation(s) in RCA: 363] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2007] [Accepted: 09/06/2007] [Indexed: 11/17/2022]
Abstract
The myocardial tissue lacks significant intrinsic regenerative capability to replace the lost cells. Therefore, the heart is a major target of research within the field of tissue engineering, which aims to replace infarcted myocardium and enhance cardiac function. The primary objective of this work was to develop a biocompatible, degradable and superelastic heart patch from poly(glycerol sebacate) (PGS). PGS was synthesised at 110, 120 and 130 degrees C by polycondensation of glycerol and sebacic acid with a mole ratio of 1:1. The investigation was focused on the mechanical and biodegrading behaviours of the developed PGS. PGS materials synthesised at 110, 120 and 130 degrees C have Young's moduli of 0.056, 0.22 and 1.2 MPa, respectively, which satisfy the mechanical requirements on the materials applied for the heart patch and 3D myocardial tissue engineering construction. Degradation assessment in phosphate buffered saline and Knockout DMEM culture medium has demonstrated that the PGS has a wide range of degradability, from being degradable in a couple of weeks to being nearly inert. The matching of physical characteristics to those of the heart, the ability to fine tune degradation rates in biologically relevant media and initial data showing biocompatibility indicate that this material has promise for cardiac tissue engineering applications.
Collapse
Affiliation(s)
- Qi-Zhi Chen
- Department of Materials, Imperial College London, Prince Consort Road, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Hsu SJ, Bouchard RR, Dumont DM, Wolf PD, Trahey GE. In vivo assessment of myocardial stiffness with acoustic radiation force impulse imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2007; 33:1706-19. [PMID: 17698282 PMCID: PMC2117626 DOI: 10.1016/j.ultrasmedbio.2007.05.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 04/16/2007] [Accepted: 05/10/2007] [Indexed: 05/04/2023]
Abstract
Acoustic radiation force impulse (ARFI) imaging has been demonstrated to be capable of visualizing variations in local stiffness within soft tissue. Recent advances in ARFI beam sequencing and parallel imaging have shortened acquisition times and lessened transducer heating to a point where ARFI acquisitions can be executed at high frame rates on commercially available diagnostic scanners. In vivo ARFI images were acquired with a linear array placed on an exposed canine heart. The electrocardiogram (ECG) was also recorded. When coregistered with the ECG, ARFI displacement images of the heart reflect the expected myocardial stiffness changes during the cardiac cycle. A radio-frequency ablation was performed on the epicardial surface of the left ventricular free wall, creating a small lesion that did not vary in stiffness during a heartbeat, though continued to move with the rest of the heart. ARFI images showed a hemispherical, stiffer region at the ablation site whose displacement magnitude and temporal variation through the cardiac cycle were less than the surrounding untreated myocardium. Sequences with radiation force pulse amplitudes set to zero were acquired to measure potential cardiac motion artifacts within the ARFI images. The results show promise for real-time cardiac ARFI imaging.
Collapse
Affiliation(s)
- Stephen J Hsu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| | | | | | | | | |
Collapse
|
44
|
Borg AN, Deepak SM, Beynon RP, Williams SG, Ray SG. A comprehensive tissue Doppler assessment of left ventricular function in patients with primary mitral regurgitation. J Am Soc Echocardiogr 2007; 20:1149-59. [PMID: 17604962 DOI: 10.1016/j.echo.2007.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Indexed: 11/28/2022]
Affiliation(s)
- Alexander N Borg
- Department of Cardiology, Wythenshawe Hospital, Manchester, United Kingdom.
| | | | | | | | | |
Collapse
|
45
|
Popović ZB, Benejam C, Bian J, Mal N, Drinko J, Lee K, Forudi F, Reeg R, Greenberg NL, Thomas JD, Penn MS. Speckle-tracking echocardiography correctly identifies segmental left ventricular dysfunction induced by scarring in a rat model of myocardial infarction. Am J Physiol Heart Circ Physiol 2007; 292:H2809-16. [PMID: 17277023 DOI: 10.1152/ajpheart.01176.2006] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Speckle-tracking echocardiography (STE) uses a two-dimensional echocardiographic image to estimate two orthogonal strain components. The aim of this study was to assess sensitivity of circumferential (Scirc) and radial (Srad) strains to infarct-induced left ventricular (LV) remodeling and scarring of the LV in a rat. To assess the relationship among Scirc, Srad, and scar size, two-dimensional echocardiographic LV short-axis images (12 MHz transducer, Vivid 7 echo machine) were collected in 34 Lewis rats 4 to 10 wk after ligation of the left anterior descending artery. Percent segmental fibrosis was assessed from histological LV cross sections stained by Masson trichrome. Ten normal rats served as echocardiographic controls. Scirc and Srad were assessed by STE. Histological data showed consistent scarring of anterior and lateral segments with variable extension to posterior and inferior segments. Both Scirc and Srad significantly decreased after myocardial infarction ( P < 0.0001 for both). As anticipated, Scirc and Srad were lowest in the infarcted segments. Multiple linear regression showed that segmental Scirc were similarly dependent on segmental fibrosis and end-systolic diameter ( P < 0.0001 for both), whereas segmental Srad measurements were more dependent on end-systolic diameter ( P < 0.0001) than on percent fibrosis ( P < 0.002). STE correctly identifies segmental LV dysfunction induced by scarring that follows myocardial infarction in rats.
Collapse
Affiliation(s)
- Zoran B Popović
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhong L, Tan RS, Ghista DN, Ng EYK, Chua LP, Kassab GS. Validation of a novel noninvasive cardiac index of left ventricular contractility in patients. Am J Physiol Heart Circ Physiol 2007; 292:H2764-72. [PMID: 17237251 DOI: 10.1152/ajpheart.00540.2006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although there are several excellent indexes of myocardial contractility, they require accurate measurement of pressure via left ventricular (LV) catheterization. Here we validate a novel noninvasive contractility index that is dependent only on lumen and wall volume of the LV chamber in patients with normal and compromised LV ejection fraction (LVEF). By analysis of the myocardial chamber as a thick-walled sphere, LV contractility index can be expressed as maximum rate of change of pressure-normalized stress (dσ*/d tmax, where σ* = σ/P and σ and P are circumferential stress and pressure, respectively). To validate this parameter, dσ*/d tmax was determined from contrast cine-ventriculography-assessed LV cavity and myocardial volumes and compared with LVEF, dP/d tmax, maximum active elastance ( Ea,max), and single-beat end-systolic elastance [ Ees(SB)] in 30 patients undergoing clinically indicated LV catheterization. Patients with different tertiles of LVEF exhibit statistically significant differences in dσ*/d tmax. There was a significant correlation between dσ*/d tmax and dP/d tmax (dσ*/d tmax = 0.0075dP/d tmax − 4.70, r = 0.88, P < 0.01), Ea,max (dσ*/d tmax = 1.20 Ea,max + 1.40, r = 0.89, P < 0.01), and Ees(SB) [dσ*/d tmax = 1.60 Ees(SB) + 1.20, r = 0.88, P < 0.01]. In 30 additional individuals, we determined sensitivity of the parameter to changes in preload (intravenous saline infusion, n = 10 subjects), afterload (sublingual glyceryl trinitrate, n = 10 subjects), and increased contractility (intravenous dobutamine, n = 10 patients). We confirmed that the index is not dependent on load but is sensitive to changes in contractility. In conclusion, dσ*/d tmax is equivalent to dP/d tmax, Ea,max, and Ees(SB) as an index of myocardial contractility and appears to be load independent. In contrast to other measures of contractility, dσ*/d tmax can be assessed with noninvasive cardiac imaging and, thereby, should have more routine clinical applicability.
Collapse
Affiliation(s)
- Liang Zhong
- Department of Cardiology, National Heart Centre, University of New South Wales-Asia, Singapore
| | | | | | | | | | | |
Collapse
|
47
|
Pacher P, Vaslin A, Benko R, Mabley JG, Liaudet L, Haskó G, Marton A, Bátkai S, Kollai M, Szabó C. A new, potent poly(ADP-ribose) polymerase inhibitor improves cardiac and vascular dysfunction associated with advanced aging. J Pharmacol Exp Ther 2004; 311:485-491. [PMID: 15213249 PMCID: PMC2527587 DOI: 10.1124/jpet.104.069658] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Increased production of reactive oxygen and nitrogen species has recently been implicated in the pathogenesis of cardiac and endothelial dysfunction associated with atherosclerosis, hypertension, and aging. Oxidant-induced cell injury triggers the activation of nuclear enzyme poly(ADP-ribose) polymerase (PARP), which in turn contributes to cardiac and vascular dysfunction in various pathophysiological conditions including diabetes, reperfusion injury, circulatory shock, and aging. Here, we investigated the effect of a new PARP inhibitor, INO-1001, on cardiac and endothelial dysfunction associated with advanced aging using Millar's new Aria pressure-volume conductance system and isolated aortic rings. Young adult (3 months old) and aging (24 months old) Fischer rats were treated for 2 months with vehicle, or the potent PARP inhibitor INO-1001. In the vehicle-treated aging animals, there was a marked reduction of both systolic and diastolic cardiac function and loss of endothelial relaxant responsiveness of aortic rings to acetylcholine. Treatment with INO-1001 improved cardiac performance in aging animals and also acetylcholine-induced, nitric oxide-mediated vascular relaxation. Thus, pharmacological inhibition of PARP may represent a novel approach to improve cardiac and vascular dysfunction associated with aging.
Collapse
Affiliation(s)
- Pál Pacher
- National Institute on Alcohol Abuse & Alcoholism, National Institutes of Health, Park Bldg. Rm. 445, 12420 Parklawn Drive, MSC-8115, Bethesda, MD 20892-8115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pacher P, Mabley JG, Liaudet L, Evgenov OV, Marton A, Haskó G, Kollai M, Szabó C. Left ventricular pressure-volume relationship in a rat model of advanced aging-associated heart failure. Am J Physiol Heart Circ Physiol 2004; 287:H2132-H2137. [PMID: 15231502 PMCID: PMC2756475 DOI: 10.1152/ajpheart.00405.2004] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aging is associated with profound changes in the structure and function of the heart. A fundamental understanding of these processes, using relevant animal models, is required for effective prevention and treatment of cardiovascular disease in the elderly. Here, we studied cardiac performance in 4- to 5-mo-old (young) and 24- to 26-mo-old (old) Fischer 344 male rats using the Millar pressure-volume (P-V) conductance catheter system. We evaluated systolic and diastolic function in vivo at different preloads, including preload recruitable stroke work (PRSW), maximal slope of the systolic pressure increment (+dP/dt), and its relation to end-diastolic volume (+dP/dt-EDV) as well as the time constant of left ventricular pressure decay, as an index of relaxation. The slope of the end-diastolic P-V relation (EDPVR), an index of left ventricular stiffness, was also calculated. Aging was associated with decrease in left ventricular systolic pressure, +dP/dt, maximal slope of the diastolic pressure decrement, +dP/dt-EDV, PRSW, ejection fraction, stroke volume, cardiac and stroke work indexes, and efficiency. In contrast, total peripheral resistance, left ventricular end-diastolic volume, left ventricular end-diastolic pressure, and EDPVR were greater in aging than in young animals. Taken together, these data suggest that advanced aging is characterized by decreased systolic performance accompanied by delayed relaxation and increased diastolic stiffness of the heart in male Fischer 344 rats. P-V analysis is a sensitive method to determine cardiac function in rats.
Collapse
Affiliation(s)
- Pál Pacher
- National Institute on Alcohol Abuse and Alcoholism, 12420 Parklawn Dr., MSC-8115, Bethesda, MD 20892-8115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Nemoto S, Razeghi P, Ishiyama M, De Freitas G, Taegtmeyer H, Carabello BA. PPAR-gamma agonist rosiglitazone ameliorates ventricular dysfunction in experimental chronic mitral regurgitation. Am J Physiol Heart Circ Physiol 2004; 288:H77-82. [PMID: 15345480 DOI: 10.1152/ajpheart.01246.2003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously we reported that the beneficial effects of beta-adrenergic blockade in chronic mitral regurgitation (MR) were in part due to induction of bradycardia, which obviously affects myocardial energy requirements. From this observation we hypothesized that part of the pathophysiology of MR may involve faulty energy substrate utilization, which in turn might lead to potentially harmful lipid accumulation as observed in other models of heart failure. To explore this hypothesis, we measured triglyceride accumulation in the myocardia of dogs with chronic MR and then attempted to enhance myocardial metabolism by chronic administration of the peroxisome proliferator-activated receptor (PPAR)-gamma agonist rosiglitazone. Cardiac tissues were obtained from three groups of dogs that included control animals, dogs with MR for 3 mo without treatment, and dogs with MR for 6 mo that were treated with rosiglitazone (8 mg/day) for the last 3 mo of observation. Hemodynamics and contractile function (end-systolic stress-strain relationship, as measured by K index) were assessed at baseline, 3 mo of MR, and 6 mo of MR (3 mo of the treatment). Lipid accumulation in MR (as indicated by oil red O staining score and TLC analysis) was marked and showed an inverse correlation with the left ventricular (LV) contractility. LV contractility was significantly restored after PPAR therapy (K index: therapy, 3.01 +/- 0.11*; 3 mo MR, 2.12 +/- 0.34; baseline, 4.01 +/- 0.29; ANOVA, P = 0.038; *P < 0.05 vs. 3 mo of MR). At the same time, therapy resulted in a marked reduction of intramyocyte lipid. We conclude that 1) chronic MR leads to intramyocyte myocardial lipid accumulation and contractile dysfunction, and 2) administration of the PPAR-gamma agonist rosiglitazone ameliorates MR-induced LV dysfunction accompanied by a decline in lipid content.
Collapse
Affiliation(s)
- Shintaro Nemoto
- Department of Medicine, Houston Veterans Affairs Medical Center, 2002 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
50
|
Pacher P, Bátkai S, Kunos G. Haemodynamic profile and responsiveness to anandamide of TRPV1 receptor knock-out mice. J Physiol 2004; 558:647-657. [PMID: 15121805 PMCID: PMC1664980 DOI: 10.1113/jphysiol.2004.064824] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2004] [Accepted: 04/29/2004] [Indexed: 11/08/2022] Open
Abstract
The endocannabinoid anandamide and cannabinoid (CB) receptors have been implicated in the hypotension in various forms of shock and in advanced liver cirrhosis. Anandamide also activates vanilloid TRPV(1) receptors on sensory nerve terminals, triggering the release of calcitonin gene-related peptide which elicits vasorelaxation in isolated blood vessels in vitro. However, the contribution of TRPV(1) receptors to the in vivo hypotensive effect of anandamide is equivocal. We compared the cardiac performance of anaesthetized TRPV(1) knockout (TRPV(1)(-/-)) mice and their wild-type (TRPV(1)(+/+)) littermates and analysed in detail the haemodynamic effects of anandamide using the Millar pressure-volume conductance catheter system. Baseline cardiovascular parameters and systolic and diastolic function at different preloads were similar in TRPV(1)(-/-) and TRPV(1)(+/+) mice. The predominant hypotensive response to bolus intravenous injections of anandamide and the associated decrease in cardiac contractility and total peripheral resistance (TPR) were similar in TRPV(1)(+/+) and TRPV(1)(-/-) mice, as was the ability of the CB(1) receptor antagonist SR141716 to completely block these effects. In TRPV(1)(+/+) mice, this hypotensive response was preceded by a transient, profound drop in cardiac contractility and heart rate and an increase in TPR, followed by a brief pressor response, effects which were unaffected by SR141716 and were absent in TRPV(1)(-/-) mice. These results indicate that mice lacking TRPV(1) receptors have a normal cardiovascular profile and their predominant cardiovascular depressor response to anandamide is mediated through CB(1) receptors. The role of TRPV(1) receptors is limited to the transient activation of the Bezold-Jarisch reflex by very high initial plasma concentrations of anandamide.
Collapse
Affiliation(s)
- Pál Pacher
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|