1
|
Luciano F, Ruggiero L, Minetti AE, Pavei G. Move less, spend more: the metabolic demands of short walking bouts. Proc Biol Sci 2024; 291:20241220. [PMID: 39410664 PMCID: PMC11521144 DOI: 10.1098/rspb.2024.1220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/03/2024] [Accepted: 08/27/2024] [Indexed: 11/01/2024] Open
Abstract
The metabolic cost of steady-state walking is well known; however, across legged animals, most walking bouts are too short to reach steady state. Here, we investigate how bout duration affects the metabolic cost of human walking with varying mechanical power, metabolic intensity and duration. Ten participants walked for 10- to 240-s bouts on a stair climber at 0.20, 0.25 and 0.36 m s-1 and on a treadmill at 1.39 m s-1. Oxygen uptake was time-integrated and divided by bout duration to get bout average uptake (V̇O2(b)). Fitting of oxygen uptake kinetics allowed calculating non-metabolic oxygen exchange during phase-I transient and, hence, non-steady-state metabolic cost (C met(b)) and efficiency. For 240-s bouts, such variables were also calculated at steady state. Across all conditions, shorter bouts had higher V̇O2(b) and C met(b), with proportionally greater non-metabolic oxygen exchange. As the bout duration increased, V̇O2(b), C met(b) and efficiency approached steady-state values. Our findings show that the time-averaged oxygen uptake and metabolic cost are greater for shorter than longer bouts: 30-s bouts consume 20-60% more oxygen than steady-state extrapolations. This is partially explained by the proportionally greater non-metabolic oxygen uptake and leads to lower efficiency for shorter bouts. Inferring metabolic cost from steady state substantially underestimates energy expenditure for short bouts.
Collapse
Affiliation(s)
- F. Luciano
- Department of Pathophysiology and Transplantation, University of Milan, Milan20133, Italy
| | - L. Ruggiero
- Department of Pathophysiology and Transplantation, University of Milan, Milan20133, Italy
- Department of Sports Science, Human Performance Research Centre, University of Konstanz, Konstanz78464, Germany
| | - A. E. Minetti
- Department of Pathophysiology and Transplantation, University of Milan, Milan20133, Italy
| | - G. Pavei
- Department of Pathophysiology and Transplantation, University of Milan, Milan20133, Italy
| |
Collapse
|
2
|
Olaizola-Rodrigo C, Castro-Abril H, Perisé-Badía I, Pancorbo L, Ochoa I, Monge R, Oliván S. Reducing Inert Materials for Optimal Cell-Cell and Cell-Matrix Interactions within Microphysiological Systems. Biomimetics (Basel) 2024; 9:262. [PMID: 38786472 PMCID: PMC11118140 DOI: 10.3390/biomimetics9050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
In the pursuit of achieving a more realistic in vitro simulation of human biological tissues, microfluidics has emerged as a promising technology. Organ-on-a-chip (OoC) devices, a product of this technology, contain miniature tissues within microfluidic chips, aiming to closely mimic the in vivo environment. However, a notable drawback is the presence of inert material between compartments, hindering complete contact between biological tissues. Current membranes, often made of PDMS or plastic materials, prevent full interaction between cell types and nutrients. Furthermore, their non-physiological mechanical properties and composition may induce unexpected cell responses. Therefore, it is essential to minimize the contact area between cells and the inert materials while simultaneously maximizing the direct contact between cells and matrices in different compartments. The main objective of this work is to minimize inert materials within the microfluidic chip while preserving proper cellular distribution. Two microfluidic devices were designed, each with a specific focus on maximizing direct cell-matrix or cell-cell interactions. The first chip, designed to increase direct cell-cell interactions, incorporates a nylon mesh with regular pores of 150 microns. The second chip minimizes interference from inert materials, thereby aiming to increase direct cell-matrix contact. It features an inert membrane with optimized macropores of 1 mm of diameter for collagen hydrogel deposition. Biological validation of both devices has been conducted through the implementation of cell migration and cell-to-cell interaction assays, as well as the development of epithelia, from isolated cells or spheroids. This endeavor contributes to the advancement of microfluidic technology, aimed at enhancing the precision and biological relevance of in vitro simulations in pursuit of more biomimetic models.
Collapse
Affiliation(s)
- Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Héctor Castro-Abril
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Laboratorio de Biomiméticos, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Ismael Perisé-Badía
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lara Pancorbo
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Monge
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
3
|
Bouhrira N, Vite A, Margulies KB. Distinct cytoskeletal regulators of mechanical memory in cardiac fibroblasts and cardiomyocytes. Basic Res Cardiol 2024; 119:277-289. [PMID: 38349539 DOI: 10.1007/s00395-023-01030-0] [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: 08/16/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 04/12/2024]
Abstract
Recognizing that cells "feel" and respond to their mechanical environment, recent studies demonstrate that many cells exhibit a phenomenon of "mechanical memory" in which features induced by prior mechanical cues persist after the mechanical stimulus has ceased. While there is a general recognition that different cell types exhibit different responses to changes in extracellular matrix stiffening, the phenomenon of mechanical memory within myocardial cell types has received little attention to date. To probe the dynamics of mechanical memory in cardiac fibroblasts (CFs) and cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs), we employed a magnetorheological elastomer (MRE) cell culture substrate with tunable and reversible stiffness spanning the range from normal to diseased myocardium. In CFs, using increased cell area and increases in α-smooth muscle actin as markers of cellular responses to matrix stiffening, we found that induction of mechanical memory required seven days of stiff priming. Both induction and maintenance of persistent CF activation were blocked with the F-actin inhibitor cytochalasin D, while inhibitors of microtubule detyrosination had no impact on CFs. In iPSC-CMs, mechanical memory was invoked after only 24 h of stiff priming. Moreover, mechanical memory induction and maintenance were microtubule-dependent in CMs with no dependence on F-actin. Overall, these results identify the distinct temporal dynamics of mechanical memory in CFs and iPSC-CMs with different cytoskeletal mediators responsible for inducing and maintaining the stiffness-activated phenotype. Due to its flexibility, this model is broadly applicable to future studies interrogating mechanotransduction and mechanical memory in the heart and might inform strategies for attenuating the impact of load-induced pathology and excess myocardial stiffness.
Collapse
Affiliation(s)
- Nesrine Bouhrira
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Alexia Vite
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Kenneth B Margulies
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Schuftan D, Kooh YKG, Guo J, Sun Y, Aryan L, Stottlemire B, Berkland C, Genin GM, Huebsch N. Dynamic control of contractile resistance to iPSC-derived micro-heart muscle arrays. J Biomed Mater Res A 2024; 112:534-548. [PMID: 37952251 PMCID: PMC10922390 DOI: 10.1002/jbm.a.37642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Many types of cardiovascular disease are linked to the mechanical forces placed on the heart. However, our understanding of how mechanical forces exactly affect the cellular biology of the heart remains incomplete. In vitro models based on cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CM) enable researchers to develop medium to high-throughput systems to study cardiac mechanobiology at the cellular level. Previous models have been developed to enable the study of mechanical forces, such as cardiac afterload. However, most of these models require exogenous extracellular matrix (ECM) to form cardiac tissues. Recently, a system was developed to simulate changes in afterload by grafting ECM-free micro-heart muscle arrays to elastomeric substrates of discrete stiffnesses. In the present study, we extended this system by combining the elastomer-grafted tissue arrays with a magnetorheological elastomeric substrate. This system allows iPSC-CM based micro-heart muscle arrays to experience dynamic changes in contractile resistance to mimic dynamically altered afterload. Acute changes in substrate stiffness led to acute changes in the calcium dynamics and contractile forces, illustrating the system's ability to dynamically elicit changes in tissue mechanics by dynamically changing contractile resistance.
Collapse
Affiliation(s)
- David Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yasaman Kargar Gaz Kooh
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingxuan Guo
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yuwen Sun
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryce Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Guy M. Genin
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Le HT, Mahara A, Fukazawa K, Nagasaki T, Yamaoka T. Widely distributable and retainable in-situ gelling material for treating myocardial infarction. Acta Biomater 2024; 176:221-233. [PMID: 38242190 DOI: 10.1016/j.actbio.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Intramyocardial hydrogel injection is a promising therapy to prevent negative remodeling following myocardial infarction (MI). In this study, we report a mechanism for in-situ gel formation without external stimulation, resulting in an injectable and tissue-retainable hydrogel for MI treatment, and investigate its therapeutic outcomes. A liquid-like polymeric solution comprising poly(3-acrylamidophenylboronic acid-co-acrylamide) (BAAm), polyvinyl alcohol (PVA), and sorbitol (S) increases the viscous modulus by reducing the pre-added sorbitol concentration is developed. This solution achieves a sol-gel transition in-vitro in heart tissue by spontaneously diffusing the sorbitol. After intramyocardial injection, the BAAm/PVA/S with lower initial viscous modulus widely spreads in the myocardium and gelate compared to a viscoelastic alginate (ALG) hydrogel and is retained longer than the BAAm/S solution. Serial echocardiogram analyses prove that injecting the BAAm/PVA/S into the hearts of subacute MI rats significantly increases the fraction shortening and ejection shortening and attenuates the expansion of systolic LV diameter for up to 21 d after injection compared to the saline injection as a control, but the ALG injection does not. In addition, histological evaluation shows that only the BAAm/PVA/S decreases the infarct size and increases the wall thickness 21 d after injection. The BAAm/PVA/S intramyocardial injection is better at restraining systolic ventricular dilatation and cardiac failure in the rat MI model than in the control groups. Our findings highlight an effective injectable hydrogel therapy for MI by optimizing injectability-dependent distribution and retention of injected material. STATEMENT OF SIGNIFICANCE: In-situ gelling material is a promising strategy for intramyocardial hydrogel injection therapy for myocardial infarction (MI). Since the sol-gel transition of reported materials is driven by external stimulation such as temperature, pH, or ultraviolet, their application in vivo remains challenging. In this study, we first reported a synthetic in-situ gelling material (BAAm/PVA/S) whose gelation is stimulated by spontaneously reducing pre-added sorbitol after contacting the heart tissue. The BAAm/PVA/S solution spreads evenly, and is retained for at least 21 d in the heart tissue. Our study demonstrated that intramyocardial injection of the BAAm/PVA/S with more extensive distribution and longer retention had better effects on preventing LV dilation and improving cardiac function after MI than that of viscoelastic ALG and saline solution. We expect that these findings provide fundamental information for the optimum design of injectable biomaterials for treating MI.
Collapse
Affiliation(s)
- Hue Thi Le
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shimmachi, Suita, Osaka 564-8565, Japan; Department of Physiology, Hanoi Medical University, Hanoi 10000, Vietnam
| | - Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shimmachi, Suita, Osaka 564-8565, Japan
| | - Kyoko Fukazawa
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shimmachi, Suita, Osaka 564-8565, Japan
| | - Takeshi Nagasaki
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shimmachi, Suita, Osaka 564-8565, Japan; Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, He 14-1, Mukai-motoori-machi, Komatsu, Ishikawa 923-0961, Japan.
| |
Collapse
|
6
|
Chen YW, Cheng PP, Yin YF, Cai H, Chen JZ, Feng MH, Guo W, Zhao P, Zhang C, Shan XL, Chen HH, Guo S, Lu Y, Xu M. Integrin αV mediated activation of myofibroblast via mechanoparacrine of transforming growth factor β1 in promoting fibrous scar formation after myocardial infarction. Biochem Biophys Res Commun 2024; 692:149360. [PMID: 38081108 DOI: 10.1016/j.bbrc.2023.149360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Myocardial infarction (MI) dramatically changes the mechanical stress, which is intensified by the fibrotic remodeling. Integrins, especially the αV subunit, mediate mechanical signal and mechanoparacrine of transforming growth factor β1 (TGF-β1) in various organ fibrosis by activating CFs into myofibroblasts (MFBs). We investigated a possible role of integrin αV mediated mechanoparacrine of TGF-β1 in MFBs activation for fibrous reparation in mice with MI. METHODS Heart samples from MI, sham, or MI plus cilengitide (14 mg/kg, specific integrin αV inhibitor) treated mice, underwent functional and morphological assessments by echocardiography, and histochemistry on 7, 14 and 28 days post-surgery. The mechanical and ultrastructural changes of the fibrous scar were further evaluated by atomic mechanics microscope (AFM), immunofluorescence, second harmonic generation (SHG) imaging, polarized light and scanning electron microscope, respectively. Hydroxyproline assay was used for total collagen content, and western blot for protein expression profile examination. Fibroblast bioactivities, including cell shape, number, Smad2/3 signal and expression of extracellular matrix (ECM) related proteins, were further evaluated by microscopic observation and immunofluorescence in polyacrylamide (PA) hydrogel with adjustable stiffness, which was re-explored in fibroblast cultured on stiff matrix after silencing of integrin αV. The content of total and free TGF-β1 was tested by enzyme-linked immunosorbent assay (ELISA) in both infarcted tissue and cell samples. RESULT Increased stiffness with heterogeneity synchronized with integrin αV and alpha smooth muscle actin (α-SMA) positive MFBs accumulation in those less mature fibrous areas. Cilengitide abruptly reduced collagen content and disrupted collagen alignment, which also decreased TGF-β1 bioavailability, Smad2/3 phosphorylation, and α-SMA expression in the fibrous area. Accordingly, fibroblast on stiff but not soft matrix exhibited obvious MFB phenotype, as evidenced by enlarged cell, hyperproliferation, well-developed α-SMA fibers, and elevated ECM related proteins, while silencing of integrin αV almost abolished this switch via attenuating paracrine of TGF-β1 and nuclear translocation of Smad2/3. CONCLUSION This study illustrated that increased tissue stiffness activates CFs into MFBs by integrin αV mediated mechanoparacrine of TGF-β1, especially in immature scar area, which ultimately promotes fibrous scar maturation.
Collapse
Affiliation(s)
- Yu-Wen Chen
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei-Pei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-Feng Yin
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Cai
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing-Zhi Chen
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming-Hui Feng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei Zhao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zhang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Li Shan
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui-Hua Chen
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuo Guo
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yi Lu
- Minhang Hospital, Fu Dan University, Shanghai, China.
| | - Ming Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
7
|
Emerson JI, Ariel P, Shi W, Conlon FL. Sex Differences in Mouse Cardiac Electrophysiology Revealed by Simultaneous Imaging of Excitation-Contraction Coupling. J Cardiovasc Dev Dis 2023; 10:479. [PMID: 38132647 PMCID: PMC10743987 DOI: 10.3390/jcdd10120479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Males and females differ in the basic anatomy and physiology of the heart. Sex differences are evident in cardiac repolarization in humans; women have longer corrected QT and JT intervals. However, the molecular mechanisms that lead to these differences are incompletely understood. Here, we present that, like in humans, sex differences in QT and JT intervals exist in mouse models; female mice had longer corrected QT and JT intervals compared with age-matched males. To further understand the molecular underpinning of these sex differences, we developed a novel technology using fluorescent confocal microscopy that allows the simultaneous visualization of action potential, Ca2+ transients, and contractions in isolated cardiomyocytes at a high temporal resolution. From this approach, we uncovered that females at baseline have increased action potential duration, decreased Ca2+ release and reuptake rates, and decreased contraction and relaxation velocities compared with males. Additionally, males had a shorter overall time from action potential onset to peak contraction. In aggregate, our studies uncovered male and female differences in excitation-contraction coupling that account for differences observed in the EKG. Overall, a better understanding of sex differences in electrophysiology is essential for equitably treating cardiac disease.
Collapse
Affiliation(s)
- James I. Emerson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Pablo Ariel
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Wei Shi
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Ahmad F, Soe S, Albon J, Errington R, Theobald P. Quantifying the microstructural and biomechanical changes in the porcine ventricles during growth and remodelling. Acta Biomater 2023; 171:166-192. [PMID: 37797709 DOI: 10.1016/j.actbio.2023.09.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Cardiac tissue growth and remodelling (G & R) occur in response to the changing physiological demands of the heart after birth. The early shift to pulmonary circulation produces an immediate increase in ventricular workload, causing microstructural and biomechanical changes that serve to maintain overall physiological homoeostasis. Such cardiac G & R continues throughout life. Quantifying the tissue's mechanical and microstructural changes because of G & R is of increasing interest, dovetailing with the emerging fields of personalised and precision solutions. This study aimed to determine equibiaxial, and non-equibiaxial extension, stress-relaxation, and the underlying microstructure of the passive porcine ventricles tissue at four time points spanning from neonatal to adulthood. The three-dimensional microstructure was investigated via two-photon excited fluorescence and second-harmonic generation microscopy on optically cleared tissues, describing the 3D orientation, rotation and dispersion of the cardiomyocytes and collagen fibrils. The results revealed that during biomechanical testing, myocardial ventricular tissue possessed non-linear, anisotropic, and viscoelastic behaviour. An increase in stiffness and viscoelasticity was noted for the left and right ventricular free walls from neonatal to adulthood. Microstructural analyses revealed concomitant increases in cardiomyocyte rotation and dispersion. This study provides baseline data, describing the biomechanical and microstructural changes in the left and right ventricular myocardial tissue during G & R, which should prove valuable to researchers in developing age-specific, constitutive models for more accurate computational simulations. STATEMENT OF SIGNIFICANCE: There is a dearth of experimental data describing the growth and remodelling of left and right ventricular tissue. The published literature is fragmented, with data reported via different experimental techniques using tissues harvested from a variety of animals, with different gender and ages. This prevents developing a continuum of data spanning birth to death, so limiting the potential that can be leveraged to aid computational modelling and simulations. In this study, equibiaxial, non-equibiaxial, and stress-relaxation data are presented, describing directional-dependent material responses. The biomechanical data is consolidated with equivalent microstructural data, an important element for the development of future material models. Combined, these data describe microstructural and biomechanical changes in the ventricles, spanning G &R from neonatal to adulthood.
Collapse
Affiliation(s)
- Faizan Ahmad
- School of Engineering, Cardiff University, UK; School of Health Sciences, Birmingham City University, UK.
| | - Shwe Soe
- FET - Engineering, Design and Mathematics, University of West of England, UK
| | - Julie Albon
- School of Optometry and Vision Sciences, Cardiff University, UK; Viva Scientia Bioimaging Laboratories, Cardiff University, UK
| | | | | |
Collapse
|
9
|
Aufan MR, Jost ZT, Miller NJ, Sharifov OF, Gupta H, Perry GJ, Wells JM, Denney TS, Lloyd SG. Electrocardiogram to Determine Mitral and Aortic Valve Opening and Closure. Cardiovasc Eng Technol 2023; 14:447-456. [PMID: 36971975 DOI: 10.1007/s13239-023-00664-4] [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: 09/19/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE Knowledge of the timing of cardiac valve opening and closing is important in cardiac physiology. The relationship between valve motion and electrocardiogram (ECG) is often assumed, however is not clearly defined. Here we investigate the accuracy of cardiac valve timing estimated using only the ECG, compared to Doppler echocardiography (DE) flow imaging as the gold standard. METHODS DE was obtained in 37 patients with simultaneous ECG recording. ECG was digitally processed and identifiable features (QRS, T, P waves) were examined as potential reference points to determine opening and closure of aortic and mitral valves, as compared to DE outflow and inflow measurement. Timing offset of the cardiac valves opening and closure between ECG features and DE was measured from derivation set (n = 19). The obtained mean offset in combination with the ECG features model was then evaluated on a validation set (n = 18). Using the same approach, additional measurement was also done for the right sided valves. RESULTS From the derivation set, we found a fixed offset of 22 ± 9 ms, 2 ± 13 ms, 90 ± 26 ms, and - 2 ± - 27 ms when comparing S to aortic valve opening, Tend to aortic valve closure, Tend to mitral valve opening, and R to mitral valve closure respectively. Application of this model to the validation set showed good estimation of aortic and mitral valve opening and closure timing value, with low model absolute error (median of the mean absolute error of the four events = 19 ms compared to the gold standard DE measurement). For the right-sided (tricuspid and pulmonic) valves in our patient set, there was considerably higher median of the mean absolute error of 42 ms for the model. CONCLUSION ECG features can be used to estimate aortic and mitral valve timings with good accuracy as compared to DE, allowing useful hemodynamic information to be derived from this easily available test.
Collapse
Affiliation(s)
- M Rifqi Aufan
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zachary T Jost
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Neal J Miller
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Oleg F Sharifov
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Himanshu Gupta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Valley Medical Group, Paramus, NJ, USA
| | - Gilbert J Perry
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Michael Wells
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Steven G Lloyd
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
10
|
Rajanathan R, Riera CVI, Pedersen TM, Staehr C, Bouzinova EV, Nyengaard JR, Thomsen MB, Bøtker HE, Matchkov VV. Hypercontractile Cardiac Phenotype in Mice with Migraine-Associated Mutation in the Na +,K +-ATPase α 2-Isoform. Cells 2023; 12:cells12081108. [PMID: 37190017 DOI: 10.3390/cells12081108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Two α-isoforms of the Na+,K+-ATPase (α1 and α2) are expressed in the cardiovascular system, and it is unclear which isoform is the preferential regulator of contractility. Mice heterozygous for the familial hemiplegic migraine type 2 (FHM2) associated mutation in the α2-isoform (G301R; α2+/G301R mice) have decreased expression of cardiac α2-isoform but elevated expression of the α1-isoform. We aimed to investigate the contribution of the α2-isoform function to the cardiac phenotype of α2+/G301R hearts. We hypothesized that α2+/G301R hearts exhibit greater contractility due to reduced expression of cardiac α2-isoform. Variables for contractility and relaxation of isolated hearts were assessed in the Langendorff system without and in the presence of ouabain (1 µM). Atrial pacing was performed to investigate rate-dependent changes. The α2+/G301R hearts displayed greater contractility than WT hearts during sinus rhythm, which was rate-dependent. The inotropic effect of ouabain was more augmented in α2+/G301R hearts than in WT hearts during sinus rhythm and atrial pacing. In conclusion, cardiac contractility was greater in α2+/G301R hearts than in WT hearts under resting conditions. The inotropic effect of ouabain was rate-independent and enhanced in α2+/G301R hearts, which was associated with increased systolic work.
Collapse
Affiliation(s)
| | - Clàudia Vilaseca I Riera
- Department of Basic Science, School of Medicine and Health Sciences, International University of Catalonia, 08195 Barcelona, Spain
| | | | - Christian Staehr
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Jens Randel Nyengaard
- Department of Clinical Medicine, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, 8000 Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Morten B Thomsen
- Biomedical Sciences, University of Copenhagen, 1168 Copenhagen, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | | |
Collapse
|
11
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
12
|
Rabbani A, Gao H, Lazarus A, Dalton D, Ge Y, Mangion K, Berry C, Husmeier D. Image-based estimation of the left ventricular cavity volume using deep learning and Gaussian process with cardio-mechanical applications. Comput Med Imaging Graph 2023; 106:102203. [PMID: 36848766 DOI: 10.1016/j.compmedimag.2023.102203] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/26/2022] [Accepted: 02/17/2023] [Indexed: 02/27/2023]
Abstract
In this investigation, an image-based method has been developed to estimate the volume of the left ventricular cavity using cardiac magnetic resonance (CMR) imaging data. Deep learning and Gaussian processes have been applied to bring the estimations closer to the cavity volumes manually extracted. CMR data from 339 patients and healthy volunteers have been used to train a stepwise regression model that can estimate the volume of the left ventricular cavity at the beginning and end of diastole. We have decreased the root mean square error (RMSE) of cavity volume estimation approximately from 13 to 8 ml compared to the common practice in the literature. Considering the RMSE of manual measurements is approximately 4 ml on the same dataset, 8 ml of error is notable for a fully automated estimation method, which needs no supervision or user-hours once it has been trained. Additionally, to demonstrate a clinically relevant application of automatically estimated volumes, we inferred the passive material properties of the myocardium given the volume estimates using a well-validated cardiac model. These material properties can be further used for patient treatment planning and diagnosis.
Collapse
Affiliation(s)
- Arash Rabbani
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom; School of Computing, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Hao Gao
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Alan Lazarus
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - David Dalton
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Yuzhang Ge
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Kenneth Mangion
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Colin Berry
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Dirk Husmeier
- School of Mathematics & Statistics, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
13
|
Computational Analysis of Cardiac Contractile Function. Curr Cardiol Rep 2022; 24:1983-1994. [PMID: 36301405 PMCID: PMC10091868 DOI: 10.1007/s11886-022-01814-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Heart failure results in the high incidence and mortality all over the world. Mechanical properties of myocardium are critical determinants of cardiac function, with regional variations in myocardial contractility demonstrated within infarcted ventricles. Quantitative assessment of cardiac contractile function is therefore critical to identify myocardial infarction for the early diagnosis and therapeutic intervention. RECENT FINDINGS Current advancement of cardiac functional assessments is in pace with the development of imaging techniques. The methods tailored to advanced imaging have been widely used in cardiac magnetic resonance, echocardiography, and optical microscopy. In this review, we introduce fundamental concepts and applications of representative methods for each imaging modality used in both fundamental research and clinical investigations. All these methods have been designed or developed to quantify time-dependent 2-dimensional (2D) or 3D cardiac mechanics, holding great potential to unravel global or regional myocardial deformation and contractile function from end-systole to end-diastole. Computational methods to assess cardiac contractile function provide a quantitative insight into the analysis of myocardial mechanics during cardiac development, injury, and remodeling.
Collapse
|
14
|
Akoumianakis I, Polkinghorne M, Antoniades C. Non-canonical WNT signalling in cardiovascular disease: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:783-797. [PMID: 35697779 PMCID: PMC9191761 DOI: 10.1038/s41569-022-00718-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
WNT signalling comprises a diverse spectrum of receptor-mediated pathways activated by a large family of WNT ligands and influencing fundamental biological processes. WNT signalling includes the β-catenin canonical pathway and the non-canonical pathways, namely the planar cell polarity and the calcium-dependent pathways. Advances over the past decade have linked non-canonical WNT signalling with key mechanisms of atherosclerosis, including oxidative stress, endothelial dysfunction, macrophage activation and vascular smooth muscle cell phenotype regulation. In addition, non-canonical WNT signalling is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and oxidative stress. Importantly, non-canonical WNT signalling activation has complex effects in adipose tissue in the context of obesity, thereby potentially linking metabolic and vascular diseases. Tissue-specific targeting of non-canonical WNT signalling might be associated with substantial risks of off-target tumorigenesis, challenging its therapeutic potential. However, novel technologies, such as monoclonal antibodies, recombinant decoy receptors, tissue-specific gene silencing with small interfering RNAs and gene editing with CRISPR-Cas9, might enable more efficient therapeutic targeting of WNT signalling in the cardiovascular system. In this Review, we summarize the components of non-canonical WNT signalling, their links with the main mechanisms of atherosclerosis, heart failure and arrhythmias, and the rationale for targeting individual components of non-canonical WNT signalling for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Murray Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Garcia-Padilla C, Lozano-Velasco E, Garcia-Lopez V, Aranega A, Franco D, Garcia-Martinez V, Lopez-Sanchez C. Comparative Analysis of Non-Coding RNA Transcriptomics in Heart Failure. Biomedicines 2022; 10:3076. [PMID: 36551832 PMCID: PMC9775550 DOI: 10.3390/biomedicines10123076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Heart failure constitutes a clinical complex syndrome with different symptomatic characteristics depending on age, sex, race and ethnicity, among others, which has become a major public health issue with an increasing prevalence. One of the most interesting tools seeking to improve prevention, diagnosis, treatment and prognosis of this pathology has focused on finding new molecular biomarkers since heart failure relies on deficient cardiac homeostasis, which is regulated by a strict gene expression. Therefore, currently, analyses of non-coding RNA transcriptomics have been oriented towards human samples. The present review develops a comparative study emphasizing the relevance of microRNAs, long non-coding RNAs and circular RNAs as potential biomarkers in heart failure. Significantly, further studies in this field of research are fundamental to supporting their widespread clinical use. In this sense, the various methodologies used by the authors should be standardized, including larger cohorts, homogeneity of the samples and uniformity of the bioinformatic pipelines used to reach stratification and statistical significance of the results. These basic adjustments could provide promising steps to designing novel strategies for clinical management of patients with heart failure.
Collapse
Affiliation(s)
- Carlos Garcia-Padilla
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
| | - Estefanía Lozano-Velasco
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Lopez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Martinez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Carmen Lopez-Sanchez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
16
|
Romaine A, Melleby AO, Alam J, Lobert VH, Lu N, Lockwood FE, Hasic A, Lunde IG, Sjaastad I, Stenmark H, Herum KM, Gullberg D, Christensen G. Integrin α11β1 and syndecan-4 dual receptor ablation attenuates cardiac hypertrophy in the pressure overloaded heart. Am J Physiol Heart Circ Physiol 2022; 322:H1057-H1071. [PMID: 35522553 DOI: 10.1152/ajpheart.00635.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the ECM in cardiac myocytes and cardiac fibroblasts, and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated amongst integrin beta 1 heterodimer-forming α subunits in response to increased afterload induced by aortic banding (AB) in wild type mice (WT). Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and post-operative analgesia, animals were administered 0.02 mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared to WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared to WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared to WT AB.
Collapse
Affiliation(s)
- Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jahedul Alam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Ning Lu
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Francesca E Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Almira Hasic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Harald Stenmark
- Institute for Cancer Research, Oslo University Hospital, Norway
| | - Kate M Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Donald Gullberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Schimmel K, Ichimura K, Reddy S, Haddad F, Spiekerkoetter E. Cardiac Fibrosis in the Pressure Overloaded Left and Right Ventricle as a Therapeutic Target. Front Cardiovasc Med 2022; 9:886553. [PMID: 35600469 PMCID: PMC9120363 DOI: 10.3389/fcvm.2022.886553] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022] Open
Abstract
Myocardial fibrosis is a remodeling process of the extracellular matrix (ECM) following cardiac stress. "Replacement fibrosis" is a term used to describe wound healing in the acute phase of an injury, such as myocardial infarction. In striking contrast, ECM remodeling following chronic pressure overload insidiously develops over time as "reactive fibrosis" leading to diffuse interstitial and perivascular collagen deposition that continuously perturbs the function of the left (L) or the right ventricle (RV). Examples for pressure-overload conditions resulting in reactive fibrosis in the LV are systemic hypertension or aortic stenosis, whereas pulmonary arterial hypertension (PAH) or congenital heart disease with right sided obstructive lesions such as pulmonary stenosis result in RV reactive fibrosis. In-depth phenotyping of cardiac fibrosis has made it increasingly clear that both forms, replacement and reactive fibrosis co-exist in various etiologies of heart failure. While the role of fibrosis in the pathogenesis of RV heart failure needs further assessment, reactive fibrosis in the LV is a pathological hallmark of adverse cardiac remodeling that is correlated with or potentially might even drive both development and progression of heart failure (HF). Further, LV reactive fibrosis predicts adverse outcome in various myocardial diseases and contributes to arrhythmias. The ability to effectively block pathological ECM remodeling of the LV is therefore an important medical need. At a cellular level, the cardiac fibroblast takes center stage in reactive fibrotic remodeling of the heart. Activation and proliferation of endogenous fibroblast populations are the major source of synthesis, secretion, and deposition of collagens in response to a variety of stimuli. Enzymes residing in the ECM are responsible for collagen maturation and cross-linking. Highly cross-linked type I collagen stiffens the ventricles and predominates over more elastic type III collagen in pressure-overloaded conditions. Research has attempted to identify pro-fibrotic drivers causing fibrotic remodeling. Single key factors such as Transforming Growth Factor β (TGFβ) have been described and subsequently targeted to test their usefulness in inhibiting fibrosis in cultured fibroblasts of the ventricles, and in animal models of cardiac fibrosis. More recently, modulation of phenotypic behaviors like inhibition of proliferating fibroblasts has emerged as a strategy to reduce pathogenic cardiac fibroblast numbers in the heart. Some studies targeting LV reactive fibrosis as outlined above have successfully led to improvements of cardiac structure and function in relevant animal models. For the RV, fibrosis research is needed to better understand the evolution and roles of fibrosis in RV failure. RV fibrosis is seen as an integral part of RV remodeling and presents at varying degrees in patients with PAH and animal models replicating the disease of RV afterload. The extent to which ECM remodeling impacts RV function and thus patient survival is less clear. In this review, we describe differences as well as common characteristics and key players in ECM remodeling of the LV vs. the RV in response to pressure overload. We review pre-clinical studies assessing the effect of anti-fibrotic drug candidates on LV and RV function and their premise for clinical testing. Finally, we discuss the mode of action, safety and efficacy of anti-fibrotic drugs currently tested for the treatment of left HF in clinical trials, which might guide development of new approaches to target right heart failure. We touch upon important considerations and knowledge gaps to be addressed for future clinical testing of anti-fibrotic cardiac therapies.
Collapse
Affiliation(s)
- Katharina Schimmel
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Kenzo Ichimura
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sushma Reddy
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Francois Haddad
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Cardiovascular Medicine, Stanford University, Stanford, CA, United States
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Edda Spiekerkoetter,
| |
Collapse
|
18
|
Tringides CM, Mooney DJ. Materials for Implantable Surface Electrode Arrays: Current Status and Future Directions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107207. [PMID: 34716730 DOI: 10.1002/adma.202107207] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/26/2021] [Indexed: 06/13/2023]
Abstract
Surface electrode arrays are mainly fabricated from rigid or elastic materials, and precisely manipulated ductile metal films, which offer limited stretchability. However, the living tissues to which they are applied are nonlinear viscoelastic materials, which can undergo significant mechanical deformation in dynamic biological environments. Further, the same arrays and compositions are often repurposed for vastly different tissues rather than optimizing the materials and mechanical properties of the implant for the target application. By first characterizing the desired biological environment, and then designing a technology for a particular organ, surface electrode arrays may be more conformable, and offer better interfaces to tissues while causing less damage. Here, the various materials used in each component of a surface electrode array are first reviewed, and then electrically active implants in three specific biological systems, the nervous system, the muscular system, and skin, are described. Finally, the fabrication of next-generation surface arrays that overcome current limitations is discussed.
Collapse
Affiliation(s)
- Christina M Tringides
- Harvard Program in Biophysics, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
19
|
Kreit J. Respiratory-Cardiovascular Interactions During Mechanical Ventilation: Physiology and Clinical Implications. Compr Physiol 2022; 12:3425-3448. [PMID: 35578946 DOI: 10.1002/cphy.c210003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Positive-pressure inspiration and positive end-expiratory pressure (PEEP) increase pleural, alveolar, lung transmural, and intra-abdominal pressure, which decrease right and left ventricular (RV; LV) preload and LV afterload and increase RV afterload. The magnitude and clinical significance of the resulting changes in ventricular function are determined by the delivered tidal volume, the total level of PEEP, the compliance of the lungs and chest wall, intravascular volume, baseline RV and LV function, and intra-abdominal pressure. In mechanically ventilated patients, the most important, adverse consequences of respiratory-cardiovascular interactions are a PEEP-induced reduction in cardiac output, systemic oxygen delivery, and blood pressure; RV dysfunction in patients with ARDS; and acute hemodynamic collapse in patients with pulmonary hypertension. On the other hand, the hemodynamic changes produced by respiratory-cardiovascular interactions can be beneficial when used to assess volume responsiveness in hypotensive patients and by reducing dyspnea and improving hypoxemia in patients with cardiogenic pulmonary edema. Thus, a thorough understanding of the physiological principles underlying respiratory-cardiovascular interactions is essential if critical care practitioners are to anticipate, recognize, manage, and utilize their hemodynamic effects. © 2022 American Physiological Society. Compr Physiol 12:1-24, 2022.
Collapse
Affiliation(s)
- John Kreit
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Lazarus A, Gao H, Luo X, Husmeier D. Improving cardio‐mechanic inference by combining in vivo strain data with ex vivo volume–pressure data. J R Stat Soc Ser C Appl Stat 2022. [DOI: 10.1111/rssc.12560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Hao Gao
- University of Glasgow GlasgowUK
| | | | | |
Collapse
|
21
|
Liu W, Nguyen-Truong M, LeBar K, Labus KM, Gray E, Ahern M, Neelakantan S, Avazmohammadi R, McGilvray KC, Puttlitz CM, Wang Z. Multiscale Contrasts Between the Right and Left Ventricle Biomechanics in Healthy Adult Sheep and Translational Implications. Front Bioeng Biotechnol 2022; 10:857638. [PMID: 35528212 PMCID: PMC9068898 DOI: 10.3389/fbioe.2022.857638] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/28/2022] [Indexed: 12/19/2022] Open
Abstract
Cardiac biomechanics play a significant role in the progression of structural heart diseases (SHDs). SHDs alter baseline myocardial biomechanics leading to single or bi-ventricular dysfunction. But therapies for left ventricle (LV) failure patients do not always work well for right ventricle (RV) failure patients. This is partly because the basic knowledge of baseline contrasts between the RV and LV biomechanics remains elusive with limited discrepant findings. The aim of the study was to investigate the multiscale contrasts between LV and RV biomechanics in large animal species. We hypothesize that the adult healthy LV and RV have distinct passive anisotropic biomechanical properties. Ex vivo biaxial tests were performed in fresh sheep hearts. Histology and immunohistochemistry were performed to measure tissue collagen. The experimental data were then fitted to a Fung type model and a structurally informed model, separately. We found that the LV was stiffer in the longitudinal (outflow tract) than circumferential direction, whereas the RV showed the opposite anisotropic behavior. The anisotropic parameter K from the Fung type model accurately captured contrasting anisotropic behaviors in the LV and RV. When comparing the elasticity in the same direction, the LV was stiffer than the RV longitudinally and the RV was stiffer than the LV circumferentially, suggesting different filling patterns of these ventricles during diastole. Results from the structurally informed model suggest potentially stiffer collagen fibers in the LV than RV, demanding further investigation. Finally, type III collagen content was correlated with the low-strain elastic moduli in both ventricles. In summary, our findings provide fundamental biomechanical differences between the chambers. These results provide valuable insights for guiding cardiac tissue engineering and regenerative studies to implement chamber-specific matrix mechanics, which is particularly critical for identifying biomechanical mechanisms of diseases or mechanical regulation of therapeutic responses. In addition, our results serve as a benchmark for image-based inverse modeling technologies to non-invasively estimate myocardial properties in the RV and LV.
Collapse
Affiliation(s)
- Wenqiang Liu
- Cardiovascular Biomechanics Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Michael Nguyen-Truong
- Cardiovascular Biomechanics Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Kristen LeBar
- Cardiovascular Biomechanics Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Kevin M. Labus
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Elisabeth Gray
- Cardiovascular Biomechanics Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Matt Ahern
- Cardiovascular Biomechanics Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Sunder Neelakantan
- Computation Cardiovascular Bioengineering Lab, Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Reza Avazmohammadi
- Computation Cardiovascular Bioengineering Lab, Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
- Computation Cardiovascular Bioengineering Lab, J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, TX, United States
| | - Kirk C. McGilvray
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
- Orthopaedic Bioengineering Research Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Christian M. Puttlitz
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
- Orthopaedic Bioengineering Research Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Zhijie Wang
- Cardiovascular Biomechanics Laboratory, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Cardiovascular Biomechanics Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
- *Correspondence: Zhijie Wang,
| |
Collapse
|
22
|
Atomic Force Microscopy (AFM) Applications in Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23073700. [PMID: 35409059 PMCID: PMC8998711 DOI: 10.3390/ijms23073700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disorder characterized by progressive replacement of cardiomyocytes by fibrofatty tissue, ventricular dilatation, cardiac dysfunction, arrhythmias, and sudden cardiac death. Interest in molecular biomechanics for these disorders is constantly growing. Atomic force microscopy (AFM) is a well-established technic to study the mechanobiology of biological samples under physiological and pathological conditions at the cellular scale. However, a review which described all the different data that can be obtained using the AFM (cell elasticity, adhesion behavior, viscoelasticity, beating force, and frequency) is still missing. In this review, we will discuss several techniques that highlight the potential of AFM to be used as a tool for assessing the biomechanics involved in ACM. Indeed, analysis of genetically mutated cells with AFM reveal abnormalities of the cytoskeleton, cell membrane structures, and defects of contractility. The higher the Young’s modulus, the stiffer the cell, and it is well known that abnormal tissue stiffness is symptomatic of a range of diseases. The cell beating force and frequency provide information during the depolarization and repolarization phases, complementary to cell electrophysiology (calcium imaging, MEA, patch clamp). In addition, original data is also presented to emphasize the unique potential of AFM as a tool to assess fibrosis in cardiac tissue.
Collapse
|
23
|
Wang H, Wisneski A, Imbrie-Moore AM, Paulsen MJ, Wang Z, Xuan Y, Lopez Hernandez H, Hironaka CE, Lucian HJ, Shin HS, Anilkumar S, Thakore AD, Farry JM, Eskandari A, Williams KM, Grady F, Wu MA, Jung J, Stapleton LM, Steele AN, Zhu Y, Woo YJ. Natural cardiac regeneration conserves native biaxial left ventricular biomechanics after myocardial infarction in neonatal rats. J Mech Behav Biomed Mater 2022; 126:105074. [PMID: 35030471 PMCID: PMC8899021 DOI: 10.1016/j.jmbbm.2022.105074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 12/23/2021] [Accepted: 01/02/2022] [Indexed: 02/03/2023]
Abstract
After myocardial infarction (MI), adult mammals exhibit scar formation, adverse left ventricular (LV) remodeling, LV stiffening, and impaired contractility, ultimately resulting in heart failure. Neonatal mammals, however, are capable of natural heart regeneration after MI. We hypothesized that neonatal cardiac regeneration conserves native biaxial LV mechanics after MI. Wistar rat neonates (1 day old, n = 46) and adults (8-10 weeks old, n = 20) underwent sham surgery or permanent left anterior descending coronary artery ligation. At 6 weeks after neonatal MI, Masson's trichrome staining revealed negligible fibrosis. Echocardiography for the neonatal MI (n = 15) and sham rats (n = 14) revealed no differences in LV wall thickness or chamber diameter, and both groups had normal ejection fraction (72.7% vs 77.5%, respectively, p = 0.1946). Biaxial tensile testing revealed similar stress-strain curves along both the circumferential and longitudinal axes across a full range of physiologic stresses and strains. The circumferential modulus (267.9 kPa vs 274.2 kPa, p = 0.7847), longitudinal modulus (269.3 kPa vs 277.1 kPa, p = 0.7435), and maximum shear stress (3.30 kPa vs 3.95 kPa, p = 0.5418) did not differ significantly between the neonatal MI and sham groups, respectively. In contrast, transmural scars were observed at 4 weeks after adult MI. Adult MI hearts (n = 7) exhibited profound LV wall thinning (p < 0.0001), chamber dilation (p = 0.0246), and LV dysfunction (ejection fraction 45.4% vs 79.7%, p < 0.0001) compared to adult sham hearts (n = 7). Adult MI hearts were significantly stiffer than adult sham hearts in both the circumferential (321.5 kPa vs 180.0 kPa, p = 0.0111) and longitudinal axes (315.4 kPa vs 172.3 kPa, p = 0.0173), and also exhibited greater maximum shear stress (14.87 kPa vs 3.23 kPa, p = 0.0162). Our study is the first to show that native biaxial LV mechanics are conserved after neonatal heart regeneration following MI, thus adding biomechanical support for the therapeutic potential of cardiac regeneration in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Andrew Wisneski
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Annabel M Imbrie-Moore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Zhongjie Wang
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Yue Xuan
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Camille E Hironaka
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Haley J Lucian
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Shreya Anilkumar
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Akshara D Thakore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Justin M Farry
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Kiah M Williams
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Frederick Grady
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Matthew A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Jinsuh Jung
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Lyndsay M Stapleton
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
Better intraoperative cardiopulmonary stability and similar postoperative results of spontaneous ventilation combined with intubation than non-intubated thoracic surgery. Gan To Kagaku Ryoho 2022; 70:559-565. [PMID: 34985733 DOI: 10.1007/s11748-021-01768-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/23/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Non-intubated spontaneous ventilation video-assisted thoracic surgery lobectomy is a well-known procedure, but there are doubts regarding its safety. To solve this problem, we developed a safe procedure for spontaneous ventilation thoracic surgery (spontaneous ventilation with intubation). This study analyzed the intraoperative parameters and postoperative results of spontaneous ventilation with intubation. METHODS Between March 11, 2020 and March 26, 2021, 38 spontaneous ventilation with intubation video-assisted thoracic surgery lobectomies were performed. We chose the first 38 non-intubated spontaneous ventilation video-assisted thoracic surgery lobectomy cases with a laryngeal mask performed in 2017 for comparison. RESULTS There were no significant differences between the non-intubated spontaneous ventilation and spontaneous ventilation with intubation groups in postoperative surgical results (surgical time: 98,7 vs. 88,1 min (p = 0.067); drainage time: 3.5 vs. 2.7 days (p = 0.194); prolonged air leak 15.7% vs. 10.5% (p = 0.5); conversion rate to relaxation: 5.2% vs. 13.1% (p = 0.237); failure of the spontaneous ventilation rate: 10.5% vs. 13.1% (p = 0.724); and morbidity: 21% vs. 13.1% (p = 0.364)) and oncological outcomes. Significantly lower lowest systolic and diastolic blood pressure (systolic, 83.1 vs 132.3 mmHg, p = 0.001; diastolic 47.8 vs. 73.4 mmHg, p = 0.0001), lowest oxygen saturation (90.3% vs 94.9%, p = 0.026), and higher maximum pCO2 level (62.5 vs 54.8 kPa, p = 0.009) were found in the non-intubated spontaneous ventilation group than in the spontaneous ventilation with intubation group. CONCLUSIONS Spontaneous ventilation with intubation is a more physiological procedure than non-intubated spontaneous ventilation in terms of intraoperative blood pressure stability and gas exchange. The surgical results were similar in the two groups.
Collapse
|
25
|
Liu X, Liu L, Zhao J, Wang H, Li Y. Mechanotransduction regulates inflammation responses of epicardial adipocytes in cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1080383. [PMID: 36589802 PMCID: PMC9800500 DOI: 10.3389/fendo.2022.1080383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is a crucial regulator in maintaining cardiovascular homeostasis by secreting various bioactive products to mediate the physiological function of the cardiovascular system. Accumulating evidence shows that adipose tissue disorders contribute to several kinds of cardiovascular disease (CVD). Furthermore, the adipose tissue would present various biological effects depending on its tissue localization and metabolic statuses, deciding the individual cardiometabolic risk. Crosstalk between adipose and myocardial tissue is involved in the pathophysiological process of arrhythmogenic right ventricular cardiomyopathy (ARVC), cardiac fibrosis, heart failure, and myocardial infarction/atherosclerosis. The abnormal distribution of adipose tissue in the heart might yield direct and/or indirect effects on cardiac function. Moreover, mechanical transduction is critical for adipocytes in differentiation, proliferation, functional maturity, and homeostasis maintenance. Therefore, understanding the features of mechanotransduction pathways in the cellular ontogeny of adipose tissue is vital for underlining the development of adipocytes involved in cardiovascular disorders, which would preliminarily contribute positive implications on a novel therapeutic invention for cardiovascular diseases. In this review, we aim to clarify the role of mechanical stress in cardiac adipocyte homeostasis and its interplay with maintaining cardiac function.
Collapse
Affiliation(s)
- Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfei Zhao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Hua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| |
Collapse
|
26
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
27
|
Villalba-Orero M, Jiménez-Riobóo RJ, Gontán N, Sanderson D, López-Olañeta M, García-Pavía P, Desco M, Lara-Pezzi E, Gómez-Gaviro MV. Assessment of myocardial viscoelasticity with Brillouin spectroscopy in myocardial infarction and aortic stenosis models. Sci Rep 2021; 11:21369. [PMID: 34725389 PMCID: PMC8560820 DOI: 10.1038/s41598-021-00661-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 02/05/2023] Open
Abstract
Heart diseases are associated with changes in the biomechanical properties of the myocardial wall. However, there is no modality available to assess myocardial stiffness directly. Brillouin microspectroscopy (mBS) is a consolidated mechanical characterization technique, applied to the study of the viscoelastic and elastic behavior of biological samples and may be a valuable tool for assessing the viscoelastic properties of the cardiac tissue. In this work, viscosity and elasticity were assessed using mBS in heart samples obtained from healthy and unhealthy mice (n = 6 per group). Speckle-tracking echocardiography (STE) was performed to evaluate heart deformation. We found that mBS was able to detect changes in stiffness in the ventricles in healthy myocardium. The right ventricle showed reduced stiffness, in agreement with its increased compliance. mBS measurements correlated strongly with STE data, highlighting the association between displacement and stiffness in myocardial regions. This correlation was lost in pathological conditions studied. The scar region in the infarcted heart presented changes in stiffness when compared to the rest of the heart, and the hypertrophied left ventricle showed increased stiffness following aortic stenosis, compared to the right ventricle. We demonstrate that mBS can be applied to determine myocardial stiffness, that measurements correlate with functional parameters and that they change with disease.
Collapse
Affiliation(s)
- María Villalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain
| | - Rafael J Jiménez-Riobóo
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Nuria Gontán
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Daniel Sanderson
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III, Madrid, Spain
| | - Marina López-Olañeta
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Pablo García-Pavía
- Hospital Puerta de Hierro Majadahonda, Madrid, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain.,Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcon, Spain
| | - Manuel Desco
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain. .,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain. .,Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), Madrid, Spain. .,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III, Madrid, Spain. .,Hospital General Universitario Gregorio Marañón, Doctor Esquerdo 46, 28007, Madrid, Spain.
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain. .,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain.
| | - Maria Victoria Gómez-Gaviro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain. .,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III, Madrid, Spain. .,Hospital General Universitario Gregorio Marañón, Doctor Esquerdo 46, 28007, Madrid, Spain.
| |
Collapse
|
28
|
Bliley JM, Vermeer MCSC, Duffy RM, Batalov I, Kramer D, Tashman JW, Shiwarski DJ, Lee A, Teplenin AS, Volkers L, Coffin B, Hoes MF, Kalmykov A, Palchesko RN, Sun Y, Jongbloed JDH, Bomer N, de Boer RA, Suurmeijer AJH, Pijnappels DA, Bolling MC, van der Meer P, Feinberg AW. Dynamic loading of human engineered heart tissue enhances contractile function and drives a desmosome-linked disease phenotype. Sci Transl Med 2021; 13:13/603/eabd1817. [PMID: 34290054 DOI: 10.1126/scitranslmed.abd1817] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/02/2021] [Indexed: 12/23/2022]
Abstract
The role that mechanical forces play in shaping the structure and function of the heart is critical to understanding heart formation and the etiology of disease but is challenging to study in patients. Engineered heart tissues (EHTs) incorporating human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have the potential to provide insight into these adaptive and maladaptive changes. However, most EHT systems cannot model both preload (stretch during chamber filling) and afterload (pressure the heart must work against to eject blood). Here, we have developed a new dynamic EHT (dyn-EHT) model that enables us to tune preload and have unconstrained contractile shortening of >10%. To do this, three-dimensional (3D) EHTs were integrated with an elastic polydimethylsiloxane strip providing mechanical preload and afterload in addition to enabling contractile force measurements based on strip bending. Our results demonstrated that dynamic loading improves the function of wild-type EHTs on the basis of the magnitude of the applied force, leading to improved alignment, conduction velocity, and contractility. For disease modeling, we used hiPSC-derived cardiomyocytes from a patient with arrhythmogenic cardiomyopathy due to mutations in the desmoplakin gene. We demonstrated that manifestation of this desmosome-linked disease state required dyn-EHT conditioning and that it could not be induced using 2D or standard 3D EHT approaches. Thus, a dynamic loading strategy is necessary to provoke the disease phenotype of diastolic lengthening, reduction of desmosome counts, and reduced contractility, which are related to primary end points of clinical disease, such as chamber thinning and reduced cardiac output.
Collapse
Affiliation(s)
- Jacqueline M Bliley
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Mathilde C S C Vermeer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Rebecca M Duffy
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ivan Batalov
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Duco Kramer
- Department of Dermatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Joshua W Tashman
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel J Shiwarski
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Andrew Lee
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Alexander S Teplenin
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Linda Volkers
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Brian Coffin
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Martijn F Hoes
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Anna Kalmykov
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Rachelle N Palchesko
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Yan Sun
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Nils Bomer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Albert J H Suurmeijer
- Department of Pathology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Daniel A Pijnappels
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Maria C Bolling
- Department of Dermatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands.
| | - Adam W Feinberg
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA. .,Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
29
|
Sahiti F, Morbach C, Henneges C, Stefenelli U, Scholz N, Cejka V, Albert J, Heuschmann PU, Ertl G, Frantz S, Angermann CE, Störk S. Dynamics of Left Ventricular Myocardial Work in Patients Hospitalized for Acute Heart Failure. J Card Fail 2021; 27:1393-1403. [PMID: 34332057 DOI: 10.1016/j.cardfail.2021.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 02/09/2023]
Abstract
BACKGROUND The left ventricular ejection fraction (LVEF) is the most commonly used measure describing pumping efficiency, but it is heavily dependent on loading conditions and therefore not well-suited to study pathophysiologic changes. The novel concept of echocardiography-derived myocardial work (MyW) overcomes this disadvantage as it is based on LV pressure-strain loops. We tracked the in-hospital changes of indices of MyW in patients admitted for acute heart failure (AHF) in relation to their recompensation status and explored the prognostic utility of MyW indices METHODS AND RESULTS: We studied 126 patients admitted for AHF (mean 73 ± 12 years, 37% female, 40% with a reduced LVEF [<40%]), providing pairs of echocardiograms obtained both on hospital admission and prior to discharge. The following MyW indices were derived: global constructive and wasted work (GCW, GWW), global work index (GWI), and global work efficiency. In patients with HF with reduced ejection fraction with decreasing N-terminal prohormone B-natriuretic peptide levels during hospitalization, the GCW and GWI improved significantly, whereas the GWW remained unchanged. In patients with HF with preserved ejection fraction, the GCW and GWI were unchanged; however, in patients with no decrease or eventual increase in N-terminal prohormone B-natriuretic peptide, we observed an increase in GWW. In all patients with AHF, higher values of GWW were associated with a higher risk of death or rehospitalization within 6 months after discharge (per 10-point increment hazard ratio 1.035, 95% confidence interval 1.005-1.065). CONCLUSIONS Our results suggest differential myocardial responses to decompensation and recompensation, depending on the HF phenotype in patients presenting with AHF. The GWW predicted the 6-month prognosis in these patients, regardless of LVEF. Future studies in larger cohorts need to confirm our results and identify determinants of short-term and longer term changes in MyW.
Collapse
Affiliation(s)
- Floran Sahiti
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Caroline Morbach
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Carsten Henneges
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
| | - Ulrich Stefenelli
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
| | - Nina Scholz
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
| | - Vladimir Cejka
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
| | - Judith Albert
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Peter U Heuschmann
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany; Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Georg Ertl
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Frantz
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Christiane E Angermann
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Störk
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany; Department of Medicine I, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
30
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
31
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
32
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
33
|
Pecchiari M, Pontikis K, Alevrakis E, Vasileiadis I, Kompoti M, Koutsoukou A. Cardiovascular Responses During Sepsis. Compr Physiol 2021; 11:1605-1652. [PMID: 33792902 DOI: 10.1002/cphy.c190044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sepsis is the life-threatening organ dysfunction arising from a dysregulated host response to infection. Although the specific mechanisms leading to organ dysfunction are still debated, impaired tissue oxygenation appears to play a major role, and concomitant hemodynamic alterations are invariably present. The hemodynamic phenotype of affected individuals is highly variable for reasons that have been partially elucidated. Indeed, each patient's circulatory condition is shaped by the complex interplay between the medical history, the volemic status, the interval from disease onset, the pathogen, the site of infection, and the attempted resuscitation. Moreover, the same hemodynamic pattern can be generated by different combinations of various pathophysiological processes, so the presence of a given hemodynamic pattern cannot be directly related to a unique cluster of alterations. Research based on endotoxin administration to healthy volunteers and animal models compensate, to an extent, for the scarcity of clinical studies on the evolution of sepsis hemodynamics. Their results, however, cannot be directly extrapolated to the clinical setting, due to fundamental differences between the septic patient, the healthy volunteer, and the experimental model. Numerous microcirculatory derangements might exist in the septic host, even in the presence of a preserved macrocirculation. This dissociation between the macro- and the microcirculation might account for the limited success of therapeutic interventions targeting typical hemodynamic parameters, such as arterial and cardiac filling pressures, and cardiac output. Finally, physiological studies point to an early contribution of cardiac dysfunction to the septic phenotype, however, our defective diagnostic tools preclude its clinical recognition. © 2021 American Physiological Society. Compr Physiol 11:1605-1652, 2021.
Collapse
Affiliation(s)
- Matteo Pecchiari
- Dipartimento di Fisiopatologia Medico Chirurgica e dei Trapianti, Università degli Studi di Milano, Milan, Italy
| | - Konstantinos Pontikis
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Emmanouil Alevrakis
- 4th Department of Pulmonary Medicine, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Ioannis Vasileiadis
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Maria Kompoti
- Intensive Care Unit, Thriassio General Hospital of Eleusis, Magoula, Greece
| | - Antonia Koutsoukou
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| |
Collapse
|
34
|
Nguyen TD, Kadri OE, Voronov RS. An Introductory Overview of Image-Based Computational Modeling in Personalized Cardiovascular Medicine. Front Bioeng Biotechnol 2020; 8:529365. [PMID: 33102452 PMCID: PMC7546862 DOI: 10.3389/fbioe.2020.529365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases account for the number one cause of deaths in the world. Part of the reason for such grim statistics is our limited understanding of the underlying mechanisms causing these devastating pathologies, which is made difficult by the invasiveness of the procedures associated with their diagnosis (e.g., inserting catheters into the coronal artery to measure blood flow to the heart). Likewise, it is also difficult to design and test assistive devices without implanting them in vivo. However, with the recent advancements made in biomedical scanning technologies and computer simulations, image-based modeling (IBM) has arisen as the next logical step in the evolution of non-invasive patient-specific cardiovascular medicine. Yet, due to its novelty, it is still relatively unknown outside of the niche field. Therefore, the goal of this manuscript is to review the current state-of-the-art and the limitations of the methods used in this area of research, as well as their applications to personalized cardiovascular investigations and treatments. Specifically, the modeling of three different physics – electrophysiology, biomechanics and hemodynamics – used in the cardiovascular IBM is discussed in the context of the physiology that each one of them describes and the mechanisms of the underlying cardiac diseases that they can provide insight into. Only the “bare-bones” of the modeling approaches are discussed in order to make this introductory material more accessible to an outside observer. Additionally, the imaging methods, the aspects of the unique cardiac anatomy derived from them, and their relation to the modeling algorithms are reviewed. Finally, conclusions are drawn about the future evolution of these methods and their potential toward revolutionizing the non-invasive diagnosis, virtual design of treatments/assistive devices, and increasing our understanding of these lethal cardiovascular diseases.
Collapse
Affiliation(s)
- Thanh Danh Nguyen
- Otto H. York Department of Chemical and Materials Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Olufemi E Kadri
- Otto H. York Department of Chemical and Materials Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States.,UC-P&G Simulation Center, University of Cincinnati, Cincinnati, OH, United States
| | - Roman S Voronov
- Otto H. York Department of Chemical and Materials Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States.,Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
35
|
Morbach C, Sahiti F, Tiffe T, Cejka V, Eichner FA, Gelbrich G, Heuschmann PU, Störk S. Myocardial work - correlation patterns and reference values from the population-based STAAB cohort study. PLoS One 2020; 15:e0239684. [PMID: 33031416 PMCID: PMC7544116 DOI: 10.1371/journal.pone.0239684] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022] Open
Abstract
Background Recently, myocardial work analysis as an echocardiographic tool to non-invasively determine LV work has been introduced and validated against invasive measurements. Based on systolic blood pressure and speckle-tracking derived longitudinal strain (GLS) during systole and isovolumic relaxation, it is considered less load-dependent than LV ejection fraction (LVEF) or GLS and to integrate information on LV active systolic and diastolic work. Objectives We aimed to establish reference values for global constructive (GCW) and global wasted work (GWW) as well as of global work index (GWI) and global work efficiency (GWE) across a wide age range and to assess the association with standard echocardiography parameters to estimate the potential additional information provided by myocardial work (MyW). Methods The Characteristics and Course of Heart Failure STAges A/B and Determinants of Progression (STAAB) cohort study carefully characterized a representative sample of the population of the City of Würzburg, Germany, aged 30–79 years. We performed myocardial work analysis using the standardized, quality-controlled transthoracic echocardiograms of all individuals lacking any cardiovascular risk factor. Results Out of 4965 participants, 779 (49±10 years, 59% women) were eligible for the present analysis. Levels of GCW, GWW, and GWE were independent of sex and body mass index, and were stable until the age of 45 years. Thereafter, we observed an upward shift to further stable values of GCW and a linear increase of GWW with advancing age, resulting in lower GWE. Age-adjusted percentiles for GCW, GWW, GWI, and GWE were derived. Higher levels of blood pressure or LV mass were associated with higher GCW, GWI, and GWW, resulting in lower GWE; higher LVEF correlated with higher GCW and GWI, but lower GWW. Higher E/e´ correlated with higher GWW, higher e´ with lower GWW. Conclusions Derived from a large sample of apparently healthy individuals from a population based-cohort, we provide age-adjusted reference values for myocardial work indices, applicable for either sex. Weak correlations with common echocardiographic parameters suggest MyW indices to potentially provide additional information, which has to be evaluated in diseased patient cohorts.
Collapse
Affiliation(s)
- Caroline Morbach
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Department of Medicine I, Cardiology, University Hospital Würzburg, Würzburg, Germany
| | - Floran Sahiti
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Department of Medicine I, Cardiology, University Hospital Würzburg, Würzburg, Germany
| | - Theresa Tiffe
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
| | - Vladimir Cejka
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
| | - Felizitas A. Eichner
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
| | - Götz Gelbrich
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Peter U. Heuschmann
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Störk
- Comprehensive Heart Failure Center, University Hospital and University of Würzburg, Würzburg, Germany
- Department of Medicine I, Cardiology, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| | | |
Collapse
|
36
|
Pseudoaneurysm Development after Free Wall Rupture Post Myocardial Infarction. J Cardiovasc Dev Dis 2020; 7:jcdd7030034. [PMID: 32906639 PMCID: PMC7570278 DOI: 10.3390/jcdd7030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 11/19/2022] Open
Abstract
Background: According to the World Health Organization, cardiovascular disease is the number one cause of death globally, claiming millions of lives each year with an increasing prevalence. Myocardial infarction (MI) makes up a large sum of these deaths each year. While MI in itself is lethal, there are several complications that can increase the morbidity and mortality of an MI, such as left ventricular wall rupture and aneurysms. Case Presentation: We present a case of an elderly male with an extensive cardiac history who presented with a non-ST segment myocardial infarction (NSTEMI) managed with percutaneous coronary intervention. Hours after, he became hemodynamically unable and was found to have a pseudoaneurysm of the left ventricle. Despite aggressive efforts, his pseudoaneurysm ruptured and he ultimately succumbed to his condition. Conclusions: Left ventricular pseudoaneurysm is usually seen after myocardial infarctions with a rupture rate of up to 45% leading to a mortality rate of about 50%. While cardiac catheterization with left ventriculography is the gold standard for diagnosis, echocardiography can also be used as an alternative. Treatment is emergent cardiac surgery but still holds a high operative risk. Therefore, patients may be medically stabilized and managed prior to ultimate surgical intervention.
Collapse
|
37
|
Li W. Biomechanics of infarcted left ventricle: a review of modelling. Biomed Eng Lett 2020; 10:387-417. [PMID: 32864174 DOI: 10.1007/s13534-020-00159-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 11/26/2022] Open
Abstract
Mathematical modelling in biomechanics of infarcted left ventricle (LV) serves as an indispensable tool for remodelling mechanism exploration, LV biomechanical property estimation and therapy assessment after myocardial infarction (MI). However, a review of mathematical modelling after MI has not been seen in the literature so far. In the paper, a systematic review of mathematical models in biomechanics of infarcted LV was established. The models include comprehensive cardiovascular system model, essential LV pressure-volume and stress-stretch models, constitutive laws for passive myocardium and scars, tension models for active myocardium, collagen fibre orientation optimization models, fibroblast and collagen fibre growth/degradation models and integrated growth-electro-mechanical model after MI. The primary idea, unique characteristics and key equations of each model were identified and extracted. Discussions on the models were provided and followed research issues on them were addressed. Considerable improvements in the cardiovascular system model, LV aneurysm model, coupled agent-based models and integrated electro-mechanical-growth LV model are encouraged. Substantial attention should be paid to new constitutive laws with respect to stress-stretch curve and strain energy function for infarcted passive myocardium, collagen fibre orientation optimization in scar, cardiac rupture and tissue damage and viscoelastic effect post-MI in the future.
Collapse
Affiliation(s)
- Wenguang Li
- School of Engineering, University of Glasgow, Glasgow, G12 8QQ UK
| |
Collapse
|
38
|
Massai D, Pisani G, Isu G, Rodriguez Ruiz A, Cerino G, Galluzzi R, Pisanu A, Tonoli A, Bignardi C, Audenino AL, Marsano A, Morbiducci U. Bioreactor Platform for Biomimetic Culture and in situ Monitoring of the Mechanical Response of in vitro Engineered Models of Cardiac Tissue. Front Bioeng Biotechnol 2020; 8:733. [PMID: 32766218 PMCID: PMC7381147 DOI: 10.3389/fbioe.2020.00733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
In the past two decades, relevant advances have been made in the generation of engineered cardiac constructs to be used as functional in vitro models for cardiac research or drug testing, and with the ultimate but still challenging goal of repairing the damaged myocardium. To support cardiac tissue generation and maturation in vitro, the application of biomimetic physical stimuli within dedicated bioreactors is crucial. In particular, cardiac-like mechanical stimulation has been demonstrated to promote development and maturation of cardiac tissue models. Here, we developed an automated bioreactor platform for tunable cyclic stretch and in situ monitoring of the mechanical response of in vitro engineered cardiac tissues. To demonstrate the bioreactor platform performance and to investigate the effects of cyclic stretch on construct maturation and contractility, we developed 3D annular cardiac tissue models based on neonatal rat cardiac cells embedded in fibrin hydrogel. The constructs were statically pre-cultured for 5 days and then exposed to 4 days of uniaxial cyclic stretch (sinusoidal waveform, 10% strain, 1 Hz) within the bioreactor. Explanatory biological tests showed that cyclic stretch promoted cardiomyocyte alignment, maintenance, and maturation, with enhanced expression of typical mature cardiac markers compared to static controls. Moreover, in situ monitoring showed increasing passive force of the constructs along the dynamic culture. Finally, only the stretched constructs were responsive to external electrical pacing with synchronous and regular contractile activity, further confirming that cyclic stretching was instrumental for their functional maturation. This study shows that the proposed bioreactor platform is a reliable device for cyclic stretch culture and in situ monitoring of the passive mechanical response of the cultured constructs. The innovative feature of acquiring passive force measurements in situ and along the culture allows monitoring the construct maturation trend without interrupting the culture, making the proposed device a powerful tool for in vitro investigation and ultimately production of functional engineered cardiac constructs.
Collapse
Affiliation(s)
- Diana Massai
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Giuseppe Pisani
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giuseppe Isu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andres Rodriguez Ruiz
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Giulia Cerino
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Renato Galluzzi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Alessia Pisanu
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andrea Tonoli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Cristina Bignardi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Alberto L Audenino
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Anna Marsano
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| |
Collapse
|
39
|
Keller BB, Kowalski WJ, Tinney JP, Tobita K, Hu N. Validating the Paradigm That Biomechanical Forces Regulate Embryonic Cardiovascular Morphogenesis and Are Fundamental in the Etiology of Congenital Heart Disease. J Cardiovasc Dev Dis 2020; 7:E23. [PMID: 32545681 PMCID: PMC7344498 DOI: 10.3390/jcdd7020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/31/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
The goal of this review is to provide a broad overview of the biomechanical maturation and regulation of vertebrate cardiovascular (CV) morphogenesis and the evidence for mechanistic relationships between function and form relevant to the origins of congenital heart disease (CHD). The embryonic heart has been investigated for over a century, initially focusing on the chick embryo due to the opportunity to isolate and investigate myocardial electromechanical maturation, the ability to directly instrument and measure normal cardiac function, intervene to alter ventricular loading conditions, and then investigate changes in functional and structural maturation to deduce mechanism. The paradigm of "Develop and validate quantitative techniques, describe normal, perturb the system, describe abnormal, then deduce mechanisms" was taught to many young investigators by Dr. Edward B. Clark and then validated by a rapidly expanding number of teams dedicated to investigate CV morphogenesis, structure-function relationships, and pathogenic mechanisms of CHD. Pioneering studies using the chick embryo model rapidly expanded into a broad range of model systems, particularly the mouse and zebrafish, to investigate the interdependent genetic and biomechanical regulation of CV morphogenesis. Several central morphogenic themes have emerged. First, CV morphogenesis is inherently dependent upon the biomechanical forces that influence cell and tissue growth and remodeling. Second, embryonic CV systems dynamically adapt to changes in biomechanical loading conditions similar to mature systems. Third, biomechanical loading conditions dynamically impact and are regulated by genetic morphogenic systems. Fourth, advanced imaging techniques coupled with computational modeling provide novel insights to validate regulatory mechanisms. Finally, insights regarding the genetic and biomechanical regulation of CV morphogenesis and adaptation are relevant to current regenerative strategies for patients with CHD.
Collapse
Affiliation(s)
- Bradley B. Keller
- Cincinnati Children’s Heart Institute, Greater Louisville and Western Kentucky Practice, Louisville, KY 40202, USA
| | - William J. Kowalski
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA;
| | - Joseph P. Tinney
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA;
| | - Kimimasa Tobita
- Department of Medical Affairs, Abiomed Japan K.K., Muromachi Higashi Mitsui Bldg, Tokyo 103-0022, Japan;
| | - Norman Hu
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84108, USA;
| |
Collapse
|
40
|
Yang P, Dong X, Zhang Y. MicroRNA profiles in plasma samples from young metabolically healthy obese patients and miRNA-21 are associated with diastolic dysfunction via TGF-β1/Smad pathway. J Clin Lab Anal 2020; 34:e23246. [PMID: 32108968 PMCID: PMC7307369 DOI: 10.1002/jcla.23246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Metabolically healthy obese patients accounts for a large part of obese population, but its clinical significance and cardiac dysfunction are often underestimated. The microRNA profiles of metabolically healthy obese patients were investigated in the study, and the selected microRNA (miRNA) based on our microarray assay will be further verified in a relatively large metabolically healthy obese population. Methods microRNA microarray was performed from six metabolically healthy obese and 6 health control blood samples. Based on the bioinformatics analysis, we further measured RT‐PCR, fibrosis markers, echocardiograms, and TGF‐β1/Smad signaling pathway in 600 metabolically healthy obese population. Results We found that miRNAs expression characteristics in metabolically healthy obese groups were markedly different from healthy control group. MiRNA‐21 was significantly increased in the samples of metabolically healthy obese patients. Besides, miRNA‐21 levels were associated with cardiac fibrosis marker. Meanwhile, higher miRNA‐21 levels were related to elevated E/E′. Besides, patients with the highest miRNA‐21 quartile showed the lowest ratio of E/A. These associations between miRNA‐21 and diastolic function parameters were independent of obesity and other confounding variables. Of note, TGF‐β1and Smad 3 were significantly upregulated while Smad 7 was downregulated according to the miRNA‐21 quartiles in metabolically healthy obese group. Conclusions We demonstrated the profiles of circulating microRNAs in metabolically healthy obese patients. Increased plasma miRNA‐21 levels were related to impaired diastolic function independent of other relevant confounding variables. MiRNA‐21 could be one of the mechanistic links between obesity and diastolic dysfunction through regulating cardiac fibrosis via TGF‐β1/Smad signaling pathway in obese hearts, which may serve as a novel target of disease intervention.
Collapse
Affiliation(s)
- Pengkang Yang
- Department of Cardiology, Xi'an No.1 hospital, Xi'an, China
| | - Xin Dong
- Department of Cardiology, Xi'an No.4 hospital, Xi'an, China.,Health Science Center Xi'an Jiaotong University, Xi'an, China
| | - Yuyang Zhang
- Department of Cardiology, Xi'an No.1 hospital, Xi'an, China
| |
Collapse
|
41
|
Simpson LJ, Reader JS, Tzima E. Mechanical Regulation of Protein Translation in the Cardiovascular System. Front Cell Dev Biol 2020; 8:34. [PMID: 32083081 PMCID: PMC7006472 DOI: 10.3389/fcell.2020.00034] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
The cardiovascular system can sense and adapt to changes in mechanical stimuli by remodeling the physical properties of the heart and blood vessels in order to maintain homeostasis. Imbalances in mechanical forces and/or impaired sensing are now not only implicated but are, in some cases, considered to be drivers for the development and progression of cardiovascular disease. There is now growing evidence to highlight the role of mechanical forces in the regulation of protein translation pathways. The canonical mechanism of protein synthesis typically involves transcription and translation. Protein translation occurs globally throughout the cell to maintain general function but localized protein synthesis allows for precise spatiotemporal control of protein translation. This Review will cover studies on the role of biomechanical stress -induced translational control in the heart (often in the context of physiological and pathological hypertrophy). We will also discuss the much less studied effects of mechanical forces in regulating protein translation in the vasculature. Understanding how the mechanical environment influences protein translational mechanisms in the cardiovascular system, will help to inform disease pathogenesis and potential areas of therapeutic intervention.
Collapse
Affiliation(s)
- Lisa J Simpson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - John S Reader
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ellie Tzima
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Jeremic N, Weber GJ, Theilen NT, Tyagi SC. Cardioprotective effects of high-intensity interval training are mediated through microRNA regulation of mitochondrial and oxidative stress pathways. J Cell Physiol 2019; 235:5229-5240. [PMID: 31823395 DOI: 10.1002/jcp.29409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
Abstract
Human studies have shown high-intensity interval training (HIIT) has beneficial cardiovascular effects and is typically more time-efficient compared with traditional endurance exercise. The main goal of this study is to show the potential molecular and functional cardiovascular benefits of HIIT compared with endurance training (ET). Three groups of mice were used including sedentary-control, ET mice, and HIIT mice groups. Results indicated ejection fraction was increased in HIIT compared with ET while fractional shortening was increased in the HIIT group compared with both groups. Blood flow of the abdominal aorta was increased in both exercise groups compared with control. Increases in cross-sectional area and mitochondrial and antioxidative markers in HIIT compared with control were observed, along with several microRNAs. These findings indicate HIIT has specific cardiac-protective effects and may be a viable alternative to traditional ET as a cardiovascular preventative medicine intervention.
Collapse
Affiliation(s)
- Nevena Jeremic
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Gregory J Weber
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Nicholas T Theilen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
43
|
Yang HX, Xu GR, Zhang C, Sun JH, Zhang Y, Song JN, Li YF, Liu Y, Li AY. The aqueous extract of Gentianella acuta improves isoproterenol‑induced myocardial fibrosis via inhibition of the TGF‑β1/Smads signaling pathway. Int J Mol Med 2019; 45:223-233. [PMID: 31939619 PMCID: PMC6889944 DOI: 10.3892/ijmm.2019.4410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/31/2019] [Indexed: 01/03/2023] Open
Abstract
Gentianella acuta (G. acuta) is one of the most commonly used herbs in Chinese Mongolian medicine for the treatment of heart disease. Previously, it was found that G. acuta ameliorated cardiac function and inhibited isoproterenol (ISO)-induced myocardial fibrosis in rats. In this study, the underlying anti-fibrotic mechanism of G. acuta was further elucidated. Histopathological changes in the heart were observed by hematoxylineosin, Masson trichrome and wheat germ agglutinin staining. Relevant molecular events were investigated using immunohistochemistry and western blotting. The results revealed that G. acuta caused improvements in myocardial injury and fibrosis. G. acuta also inhibited collagens I and III and α-smooth muscle actin production in heart tissue. G. acuta downregulated the expression of transforming growth factor β1 (TGF-β1) and notably inhibited the levels of phosphorylation of TGF-β receptors I and II. Furthermore, G. acuta caused downregulation of the intracellular mothers against decapentaplegic homolog (Smads)2 and 4 expression and inhibited Smads2 and 3 phosphorylation. The results further demonstrated that the mechanism underlying anti-myocardial fibrosis effects of G. acuta was based upon the suppression of the TGF-β1/Smads signaling pathway. Therefore, G. acuta may be a potential therapeutic agent for ameliorating myocardial fibrosis.
Collapse
Affiliation(s)
- Hong-Xia Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Geng-Rui Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Chuang Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jia-Huan Sun
- Department of Medical Laboratory Science, College of Integration of Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jun-Na Song
- Department of Medicinal Plant, College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yun-Feng Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Ai-Ying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
44
|
Abbaszadeh S, Garjani A, Nazemiyeh H, Ayadi S, Mohajer Milani M, Soraya H. Hydroalcoholic Extract from Rhizomes of Cynodon dactylon Improve Hemodynamic and Electrocardiogram Parameters in Myocardial Infarction in Rats. PHARMACEUTICAL SCIENCES 2019. [DOI: 10.15171/ps.2019.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Cynodon dactylon is a herbal medicine of interest in Iranian traditional medicine, which is used in cardiovascular diseases such as atherosclerosis and heart failure. The purpose of this study was to evaluate the effects of total extract of C. dactylon rhizomes on myocardial infarction and on post myocardial infarction (MI) heart tissue injuries. Methods: Isoproterenol (100 mg/kg) was injected subcutaneously for two consecutive days for induction of MI in rats and C. dactylon extract was administered orally twice daily started before isoproterenol injection for 4 consecutive days. Results: Histopathological analysis showed a marked increase in myocardial necrosis in rats with MI (p<0.001). Treatment with C. dactylon (200 mg/kg) significantly (P<0.05) decreased myocardial necrosis. Hemodynamic variables were significantly suppressed in MI group and treatment with C. dactylon improved the hemodynamic parameters (P<0.05). Our electrocardiogram analysis demonstrated that C. dactylon with all doses increased R-Amplitude and R-R Interval (p<0.05, p<0.01) which were suppressed in MI group. Furthermore in treated groups with 100 and 200 mg/kg, P-R interval was also significantly increased in compared to MI group. Conclusion: This study demonstrated that C. dactylon can improve hemodynamic and electrocardiogram parameters in isoproterenol-induced myocardial infarction and thereby suggest that it can be used as a cardioprotective agent in myocardial infarction.
Collapse
Affiliation(s)
- Samin Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Garjani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Nazemiyeh
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Ayadi
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Majid Mohajer Milani
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
45
|
Oglesby M, Escobedo D, Escobar GP, Fatemifar F, Sako EY, Bailey SR, Han HC, Feldman MD. Trabecular cutting: a novel surgical therapy to increase diastolic compliance. J Appl Physiol (1985) 2019; 127:457-463. [PMID: 31219774 DOI: 10.1152/japplphysiol.00087.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common cause of hospital admission in patients over 65 yr old and has high mortality. HFpEF is characterized by left ventricular (LV) hypertrophy that reduces compliance. Current HFpEF therapies control symptoms, but no existing medications or therapies can sustainably increase LV compliance. LV trabeculae develop hypertrophy and fibrosis that contribute to reduced LV compliance. This study expands our previous results in ex vivo human hearts to show that severing LV trabeculae increases diastolic compliance in an ex vivo working rabbit heart model. Trabecular cutting was performed in ex vivo rabbit hearts set up in a working heart perfusion system perfused with oxygenated Krebs-Henseleit buffer. A hook was inserted in the LV to cut trabeculae. End-systolic and end-diastolic pressure-volume relationships during transient preload reduction were recorded using an admittance catheter in the following three groups: control (no cutting; n = 9), mild cutting (15 cuts; n = 5), and aggressive cutting (30 cuts; n = 5). In a second experiment, each heart served as its own control. Hemodynamic data were recorded before and after trabecular cutting (n = 10) or sham cutting (n = 5) within the same heart. In the first experiments, trabecular cutting did not affect systolic function (P > 0.05) but significantly increased overall diastolic compliance (P = 0.009). Greater compliance was seen as trabecular cutting increased (P = 0.002, r2 = 0.435). In the second experiment, significant increases in systolic function (P = 0.048) and diastolic compliance (P = 0.002) were seen after trabecular cutting compared with baseline. In conclusion, trabecular cutting significantly increases diastolic compliance without reducing systolic function.NEW & NOTEWORTHY We postulate that, in mammalian hearts, free-running trabeculae carneae exist to provide tensile support to the left ventricle and minimize diastolic wall stress. Because of hypertrophy and fibrosis of trabeculae in patients with left ventricular hypertrophy, this supportive role can become pathologic, worsening diastolic compliance. We demonstrate a novel operation involving cutting trabeculae as a method to acutely increase diastolic compliance in patients presenting with heart failure and diastolic dysfunction to improve their left ventricle compliance.
Collapse
Affiliation(s)
- Meagan Oglesby
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Danny Escobedo
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Gladys Patricia Escobar
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Fatemeh Fatemifar
- Department of Mechanical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Edward Y Sako
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Steven R Bailey
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Hai-Chao Han
- Department of Mechanical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Marc D Feldman
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
46
|
Wang H, Wisneski A, Paulsen MJ, Imbrie-Moore A, Wang Z, Xuan Y, Hernandez HL, Lucian HJ, Eskandari A, Thakore AD, Farry JM, Hironaka CE, von Bornstaedt D, Steele AN, Stapleton LM, Williams KM, Wu MA, MacArthur JW, Woo YJ. Bioengineered analog of stromal cell-derived factor 1α preserves the biaxial mechanical properties of native myocardium after infarction. J Mech Behav Biomed Mater 2019; 96:165-171. [PMID: 31035067 DOI: 10.1016/j.jmbbm.2019.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/04/2019] [Accepted: 04/11/2019] [Indexed: 01/08/2023]
Abstract
Adverse remodeling of the left ventricle (LV) after myocardial infarction (MI) results in abnormal tissue biomechanics and impaired cardiac function, often leading to heart failure. We hypothesized that intramyocardial delivery of engineered stromal cell-derived factor 1α analog (ESA), our previously-developed supra-efficient pro-angiogenic chemokine, preserves biaxial LV mechanical properties after MI. Male Wistar rats (n = 45) underwent sham surgery (n = 15) or permanent left anterior descending coronary artery ligation. Rats sustaining MI were randomized for intramyocardial injections of either saline (100 μL, n = 15) or ESA (6 μg/kg, n = 15), delivered at four standardized borderzone sites. After 4 weeks, echocardiography was performed, and the hearts were explanted. Tensile testing of the anterolateral LV wall was performed using a displacement-controlled biaxial load frame, and modulus was determined after constitutive modeling. At 4 weeks post-MI, compared to saline controls, ESA-treated hearts had greater wall thickness (1.68 ± 0.05 mm vs 1.42 ± 0.08 mm, p = 0.008), smaller end-diastolic LV internal dimension (6.88 ± 0.29 mm vs 7.69 ± 0.22 mm, p = 0.044), and improved ejection fraction (62.8 ± 3.0% vs 49.4 ± 4.5%, p = 0.014). Histologic analysis revealed significantly reduced infarct size for ESA-treated hearts compared to saline controls (29.4 ± 2.9% vs 41.6 ± 3.1%, p = 0.021). Infarcted hearts treated with ESA exhibited decreased modulus compared to those treated with saline in both the circumferential (211.5 ± 6.9 kPa vs 264.3 ± 12.5 kPa, p = 0.001) and longitudinal axes (194.5 ± 6.5 kPa vs 258.1 ± 14.4 kPa, p < 0.001). In both principal directions, ESA-treated infarcted hearts possessed similar tissue compliance as sham non-infarcted hearts. Overall, intramyocardial ESA therapy improves post-MI ventricular remodeling and function, reduces infarct size, and preserves native LV biaxial mechanical properties.
Collapse
Affiliation(s)
- Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Andrew Wisneski
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Annabel Imbrie-Moore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Zhongjie Wang
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Yue Xuan
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Haley J Lucian
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Akshara D Thakore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Justin M Farry
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Camille E Hironaka
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Lyndsay M Stapleton
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Kiah M Williams
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Matthew A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - John W MacArthur
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
47
|
Fatemifar F, Feldman MD, Oglesby M, Han HC. Comparison of Biomechanical Properties and Microstructure of Trabeculae Carneae, Papillary Muscles, and Myocardium in the Human Heart. J Biomech Eng 2019; 141:021007. [PMID: 30418486 PMCID: PMC6298537 DOI: 10.1115/1.4041966] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/28/2018] [Indexed: 12/13/2022]
Abstract
Trabeculae carneae account for a significant portion of human ventricular mass, despite being considered embryologic remnants. Recent studies have found trabeculae hypertrophy and fibrosis in hypertrophied left ventricles with various pathological conditions. The objective of this study was to investigate the passive mechanical properties and microstructural characteristics of trabeculae carneae and papillary muscles compared to the myocardium in human hearts. Uniaxial tensile tests were performed on samples of trabeculae carneae and myocardium strips, while biaxial tensile tests were performed on samples of papillary muscles and myocardium sheets. The experimental data were fitted with a Fung-type strain energy function and material coefficients were determined. The secant moduli at given diastolic stress and strain levels were determined and compared among the tissues. Following the mechanical testing, histology examinations were performed to investigate the microstructural characteristics of the tissues. Our results demonstrated that the trabeculae carneae were significantly stiffer (Secant modulus SM2 = 80.06 ± 10.04 KPa) and had higher collagen content (16.10 ± 3.80%) than the myocardium (SM2 = 55.14 ± 20.49 KPa, collagen content = 10.06 ± 4.15%) in the left ventricle. The results of this study improve our understanding of the contribution of trabeculae carneae to left ventricular compliance and will be useful for building accurate computational models of the human heart.
Collapse
Affiliation(s)
- Fatemeh Fatemifar
- Department of Mechanical Engineering,
University of Texas at San Antonio,
San Antonio, TX 78249
| | - Marc D. Feldman
- Department of Medicine,
University of Texas Health Science
Center at San Antonio,
San Antonio, TX 78229
| | - Meagan Oglesby
- Department of Medicine,
University of Texas Health Science
Center at San Antonio,
San Antonio, TX 78229
| | - Hai-Chao Han
- Department of Mechanical Engineering,
University of Texas at San Antonio,
San Antonio, TX 78249
e-mail:
| |
Collapse
|
48
|
Farré N, Jorba I, Torres M, Falcones B, Martí-Almor J, Farré R, Almendros I, Navajas D. Passive Stiffness of Left Ventricular Myocardial Tissue Is Reduced by Ovariectomy in a Post-menopause Mouse Model. Front Physiol 2018; 9:1545. [PMID: 30455648 PMCID: PMC6230582 DOI: 10.3389/fphys.2018.01545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Heart failure (HF) – a very prevalent disease with high morbidity and mortality – usually presents with diastolic dysfunction. Although post-menopause women are at increased risk of HF and diastolic dysfunction, poor attention has been paid to clinically and experimentally investigate this group of patients. Specifically, whether myocardial stiffness is affected by menopause is unknown. Aim: To investigate whether loss of female sexual hormones modifies the Young’s modulus (E) of left ventricular (LV) myocardial tissue in a mouse model of menopause induced by ovariectomy (OVX). Methods: After 6 months of bilateral OVX, eight mice were sacrificed, fresh LV myocardial strips were prepared (∼8 × 1 × 1 mm), and their passive stress–stretch relationship was measured. E was computed by exponential fitting of the stress–stretch relationship. Subsequently, to assess the relative role of cellular and extracellular matrix components in determining OVX-induced changes in E, the tissues strips were decellularized and subjected to the same stretching protocol to measure E. A control group of eight sham-OVX mice was simultaneously studied. Results: E (kPa; m ± SE) in OVX mice was ∼twofold lower than in controls (11.7 ± 1.8 and 22.1 ± 4.4, respectively; p < 0.05). No significant difference between groups was found in E of the decellularized tissue (31.4 ± 12.05 and 40.9 ± 11.5, respectively; p = 0.58). Conclusion: Loss of female sexual hormones in an OVX model induces a reduction in the passive stiffness of myocardial tissue, suggesting that active relaxation should play a counterbalancing role in diastolic dysfunction in post-menopausal women with HF.
Collapse
Affiliation(s)
- Núria Farré
- Heart Failure Unit, Department of Cardiology, Hospital del Mar, Barcelona, Spain.,Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignasi Jorba
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia - The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marta Torres
- Department of Pneumology, Hospital Clinic Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Bryan Falcones
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Julio Martí-Almor
- Heart Failure Unit, Department of Cardiology, Hospital del Mar, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain
| | - Isaac Almendros
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia - The Barcelona Institute of Science and Technology, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| |
Collapse
|
49
|
Mahmood SS, Pinsky MR. Heart-lung interactions during mechanical ventilation: the basics. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:349. [PMID: 30370276 DOI: 10.21037/atm.2018.04.29] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The hemodynamic effects of mechanical ventilation can be grouped into three clinically relevant concepts. First, since spontaneous ventilation is exercise. In patients increased work of breathing, initiation of mechanical ventilatory support may improve O2 delivery because the work of breathing is reduced. Second, changes in lung volume alter autonomic tone, pulmonary vascular resistance, and at high lung volumes compress the heart in the cardiac fossa similarly to cardiac tamponade. As lung volume increases so does the pressure difference between airway and pleural pressure. When this pressure difference exceeds pulmonary artery pressure, pulmonary vessels collapse as they pass form the pulmonary arteries into the alveolar space increasing pulmonary vascular resistance. Hyperinflation increases pulmonary vascular resistance impeding right ventricular ejection. Anything that over distends lung units will increase their vascular resistance, and if occurring globally throughout the lung, increase pulmonary vascular resistance. Decreases in end-expiratory lung volume cause alveolar collapse increases pulmonary vasomotor tone by the process of hypoxic pulmonary vasoconstriction. Recruitment maneuvers that restore alveolar oxygenation without over distention will reduce pulmonary artery pressure. Third, positive-pressure ventilation increases intrathoracic pressure. Since diaphragmatic descent increases intra-abdominal pressure, the decrease in the pressure gradient for venous return is less than would otherwise occur if the only change were an increase in right atrial pressure. However, in hypovolemic states, it can induce profound decreases in venous return. Increases in intrathoracic pressure decreases left ventricular afterload and will augment left ventricular ejection. In patients with hypervolemic heart failure, this afterload reducing effect can result in improved left ventricular ejection, increased cardiac output and reduced myocardial O2 demand. This brief review will focus primarily on mechanical ventilation and intrathoracic pressure as they affect right and left ventricular function and cardiac output.
Collapse
Affiliation(s)
- Syed S Mahmood
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Pinsky
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
50
|
Farré N, Otero J, Falcones B, Torres M, Jorba I, Gozal D, Almendros I, Farré R, Navajas D. Intermittent Hypoxia Mimicking Sleep Apnea Increases Passive Stiffness of Myocardial Extracellular Matrix. A Multiscale Study. Front Physiol 2018; 9:1143. [PMID: 30158879 PMCID: PMC6104184 DOI: 10.3389/fphys.2018.01143] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Tissue hypoxia-reoxygenation characterizes obstructive sleep apnea (OSA), a very prevalent respiratory disease associated with increased cardiovascular morbidity and mortality. Experimental studies indicate that intermittent hypoxia (IH) mimicking OSA induces oxidative stress and inflammation in heart tissue at the cell and molecular levels. However, it remains unclear whether IH modifies the passive stiffness of the cardiac tissue extracellular matrix (ECM). Aim: To investigate multiscale changes of stiffness induced by chronic IH in the ECM of left ventricular (LV) myocardium in a murine model of OSA. Methods: Two-month and 18-month old mice (N = 10 each) were subjected to IH (20% O2 40 s-6% O2 20 s) for 6 weeks (6 h/day). Corresponding control groups for each age were kept under normoxia. Fresh LV myocardial strips (∼7 mm × 1 mm × 1 mm) were prepared, and their ECM was obtained by decellularization. Myocardium ECM macroscale mechanics were measured by performing uniaxial stress-strain tensile tests. Strip macroscale stiffness was assessed as the stress value (σ) measured at 0.2 strain and Young's modulus (EM) computed at 0.2 strain by fitting Fung's constitutive model to the stress-strain relationship. ECM stiffness was characterized at the microscale as the Young's modulus (Em) measured in decellularized tissue slices (∼12 μm tick) by atomic force microscopy. Results: Intermittent hypoxia induced a ∼1.5-fold increase in σ (p < 0.001) and a ∼2.5-fold increase in EM (p < 0.001) of young mice as compared with normoxic controls. In contrast, no significant differences emerged in Em among IH-exposed and normoxic mice. Moreover, the mechanical effects of IH on myocardial ECM were similar in young and aged mice. Conclusion: The marked IH-induced increases in macroscale stiffness of LV myocardium ECM suggests that the ECM plays a role in the cardiac dysfunction induced by OSA. Furthermore, absence of any significant effects of IH on the microscale ECM stiffness suggests that the significant increases in macroscale stiffening are primarily mediated by 3D structural ECM remodeling.
Collapse
Affiliation(s)
- Núria Farré
- Heart Failure Unit, Department of Cardiology, Hospital del Mar, Barcelona, Spain.,Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jorge Otero
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Bryan Falcones
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Marta Torres
- CIBER de Enfermedades Respiratorias, Madrid, Spain.,Sleep Lab, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Ignasi Jorba
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - David Gozal
- Department of Child Health, University of Missouri School of Medicine, Columbia, MO, United States
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|