1
|
Siddique AB, Williams KA, Swami NS. Nanogrooved Elastomeric Diaphragm Arrays for Assessment of Cardiomyocytes under Synergistic Effects of Circular Mechanical Stimuli and Electrical Conductivity to Enhance Intercellular Communication. ACS Biomater Sci Eng 2025; 11:672-681. [PMID: 39679605 PMCID: PMC11733923 DOI: 10.1021/acsbiomaterials.4c01298] [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: 07/14/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
Cardiovascular diseases remain the leading cause of mortality, necessitating advancements in in vitro cardiac tissue engineering platforms for improved disease modeling, drug screening, and regenerative therapies. The chief challenge to recapitulating the beating behavior of cardiomyocytes is creation of the circular stress profile experienced by hollow organs in the natural heart due to filling pressure and integrated strategies for intercellular communication to promote cell-to-cell connections. We present a platform featuring addressable arrays of nanogrooved polydimethylsiloxane (PDMS) diaphragms for cell alignment and circular mechanical stimulation, with embedded silver nanowires (AgNWs) for electrical cues, so that cardiomyocyte functionality can be assessed under these synergistic influences. Central to our innovation is a two-layer PDMS diaphragm design that electrically isolates the liquid metal (EGaIn) strain sensor in the bottom layer to enable detection and control of mechanical stimulation from conductive portions of embedded AgNWs in the top layer that supports cardiomyocyte culture and communication. In this manner, through localized detection and control of the circular mechanical stimulation, the essential role of multiaxial stretching on cardiomyocyte function is elucidated based on their contractility, sarcomere length, and connexin-43 expression. This in vitro platform can potentially transform cardiac tissue engineering, drug screening, and precision medicine approaches.
Collapse
Affiliation(s)
- Abdullah-Bin Siddique
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Keith A. Williams
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Nathan S. Swami
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
- Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
2
|
Romanelli G, Villarreal L, Espasandín C, Benech JC. Diabetes induces modifications in costameric proteins and increases cardiomyocyte stiffness. Am J Physiol Cell Physiol 2024; 327:C1263-C1273. [PMID: 39374079 DOI: 10.1152/ajpcell.00273.2024] [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: 04/26/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024]
Abstract
Several studies have demonstrated that diabetes mellitus can increase the risk of cardiovascular disease and remains the principal cause of death in these patients. Costameres connect the sarcolemma with the cytoskeleton and extracellular matrix, facilitating the transmission of mechanical forces and cell signaling. They are related to cardiac physiology because individual cardiac cells are connected by intercalated discs that synchronize muscle contraction. Diabetes impacts the nanomechanical properties of cardiomyocytes, resulting in increased cellular and left ventricular stiffness, as evidenced in clinical studies of these patients. The question of whether costameric proteins are affected by diabetes in the heart has not been studied. This work analyzes whether type 1 diabetes mellitus (T1DM) modifies the costameric proteins and coincidentally changes the cellular mechanics in the same cardiomyocytes. The samples were analyzed by immunotechniques using laser confocal microscopy. Significant statistical differences were found in the spatial arrangement of the costameric proteins. However, these differences are not due to their expression. Atomic force microscopy was used to compare intrinsic cellular stiffness between diabetic and normal cardiomyocytes and obtain the first elasticity map sections of diabetic living cardiomyocytes. Data obtained demonstrated that diabetic cardiomyocytes had higher stiffness than control. The present work shows experimental evidence that intracellular changes related to cell-cell and cell-extracellular matrix communication occur, which could be related to cardiac pathogenic mechanisms. These changes could contribute to alterations in the mechanical and electrical properties of cardiomyocytes and, consequently, to diabetic cardiomyopathy.NEW & NOTEWORTHY The structural organization of cardiomyocyte proteins is critical for their efficient functioning as a contractile unit in the heart. This work shows that diabetes mellitus induces significant changes in the spatial organization of costamere proteins, t tubules, and intercalated discs. We obtained the first elasticity map sections of living diabetic cardiomyocytes. The results show statistical differences in the map sections of diabetic and control cardiomyocytes, with diabetic cardiomyocytes being stiffer than normal ones.
Collapse
Affiliation(s)
- Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Lihuén Villarreal
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Camila Espasandín
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Juan Claudio Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
3
|
Masuda A, Kurashina Y, Tani H, Soma Y, Muramatsu J, Itai S, Tohyama S, Onoe H. Maturation of Human iPSC-Derived Cardiac Microfiber with Electrical Stimulation Device. Adv Healthc Mater 2024; 13:e2303477. [PMID: 38768494 DOI: 10.1002/adhm.202303477] [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: 10/11/2023] [Revised: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Here an electrical stimulation system is described for maturing microfiber-shaped cardiac tissue (cardiac microfibers, CMFs). The system enables stable culturing of CMFs with electrical stimulation by placing the tissue between electrodes. The electrical stimulation device provides an electric field covering whole CMFs within the stimulation area and can control the beating of the cardiac microfibers. In addition, CMFs under electrical stimulation with different frequencies are examined to evaluate the maturation levels by their sarcomere lengths, electrophysiological characteristics, and gene expression. Sarcomere elongation (14% increase compared to control) is observed at day 10, and a significant upregulation of electrodynamic properties such as gap junction protein alpha 1 (GJA1) and potassium inwardly rectifying channel subfamily J member 2 (KCNJ2) (maximum fourfold increase compared to control) is observed at day 30. These results suggest that electrically stimulated cultures can accelerate the maturation of microfiber-shaped cardiac tissues compared to those without electrical stimulation. This model will contribute to the pathological research of unexplained cardiac diseases and pharmacologic testing by stably constructing matured CMFs.
Collapse
Affiliation(s)
- Akari Masuda
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Yuta Kurashina
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo, 144-0041, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo, 144-0041, Japan
| | - Jumpei Muramatsu
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Shun Itai
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
- Division of Medical Science, Graduate School of Biomedical Engineering, Tohoku University, 1-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo, 144-0041, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| |
Collapse
|
4
|
Xu F, Jin H, Liu L, Yang Y, Cen J, Wu Y, Chen S, Sun D. Architecture design and advanced manufacturing of heart-on-a-chip: scaffolds, stimulation and sensors. MICROSYSTEMS & NANOENGINEERING 2024; 10:96. [PMID: 39006908 PMCID: PMC11239895 DOI: 10.1038/s41378-024-00692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 07/16/2024]
Abstract
Heart-on-a-chip (HoC) has emerged as a highly efficient, cost-effective device for the development of engineered cardiac tissue, facilitating high-throughput testing in drug development and clinical treatment. HoC is primarily used to create a biomimetic microphysiological environment conducive to fostering the maturation of cardiac tissue and to gather information regarding the real-time condition of cardiac tissue. The development of architectural design and advanced manufacturing for these "3S" components, scaffolds, stimulation, and sensors is essential for improving the maturity of cardiac tissue cultivated on-chip, as well as the precision and accuracy of tissue states. In this review, the typical structures and manufacturing technologies of the "3S" components are summarized. The design and manufacturing suggestions for each component are proposed. Furthermore, key challenges and future perspectives of HoC platforms with integrated "3S" components are discussed. Architecture design concepts of scaffolds, stimulation and sensors in chips.
Collapse
Affiliation(s)
- Feng Xu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Hang Jin
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Lingling Liu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Yuanyuan Yang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Jianzheng Cen
- Guangdong Provincial People’s Hospital, Guangzhou, 510080 China
| | - Yaobin Wu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Songyue Chen
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Daoheng Sun
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| |
Collapse
|
5
|
Coppini A, Falconieri A, Mualem O, Nasrin SR, Roudon M, Saper G, Hess H, Kakugo A, Raffa V, Shefi O. Can repetitive mechanical motion cause structural damage to axons? Front Mol Neurosci 2024; 17:1371738. [PMID: 38912175 PMCID: PMC11191579 DOI: 10.3389/fnmol.2024.1371738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Biological structures have evolved to very efficiently generate, transmit, and withstand mechanical forces. These biological examples have inspired mechanical engineers for centuries and led to the development of critical insights and concepts. However, progress in mechanical engineering also raises new questions about biological structures. The past decades have seen the increasing study of failure of engineered structures due to repetitive loading, and its origin in processes such as materials fatigue. Repetitive loading is also experienced by some neurons, for example in the peripheral nervous system. This perspective, after briefly introducing the engineering concept of mechanical fatigue, aims to discuss the potential effects based on our knowledge of cellular responses to mechanical stresses. A particular focus of our discussion are the effects of mechanical stress on axons and their cytoskeletal structures. Furthermore, we highlight the difficulty of imaging these structures and the promise of new microscopy techniques. The identification of repair mechanisms and paradigms underlying long-term stability is an exciting and emerging topic in biology as well as a potential source of inspiration for engineers.
Collapse
Affiliation(s)
| | | | - Oz Mualem
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| | - Syeda Rubaiya Nasrin
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | - Marine Roudon
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Gadiel Saper
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Akira Kakugo
- Graduate School of Science, Division of Physics and Astronomy, Kyoto University, Kyoto, Japan
| | | | - Orit Shefi
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
6
|
Casarella S, Ferla F, Di Francesco D, Canciani E, Rizzi M, Boccafoschi F. Focal Adhesion's Role in Cardiomyocytes Function: From Cardiomyogenesis to Mechanotransduction. Cells 2024; 13:664. [PMID: 38667279 PMCID: PMC11049660 DOI: 10.3390/cells13080664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Mechanotransduction refers to the ability of cells to sense mechanical stimuli and convert them into biochemical signals. In this context, the key players are focal adhesions (FAs): multiprotein complexes that link intracellular actin bundles and the extracellular matrix (ECM). FAs are involved in cellular adhesion, growth, differentiation, gene expression, migration, communication, force transmission, and contractility. Focal adhesion signaling molecules, including Focal Adhesion Kinase (FAK), integrins, vinculin, and paxillin, also play pivotal roles in cardiomyogenesis, impacting cell proliferation and heart tube looping. In fact, cardiomyocytes sense ECM stiffness through integrins, modulating signaling pathways like PI3K/AKT and Wnt/β-catenin. Moreover, FAK/Src complex activation mediates cardiac hypertrophic growth and survival signaling in response to mechanical loads. This review provides an overview of the molecular and mechanical mechanisms underlying the crosstalk between FAs and cardiac differentiation, as well as the role of FA-mediated mechanotransduction in guiding cardiac muscle responses to mechanical stimuli.
Collapse
Affiliation(s)
- Simona Casarella
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Federica Ferla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Dalila Di Francesco
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Elena Canciani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Manuela Rizzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| |
Collapse
|
7
|
Khanna A, Oropeza BP, Huang NF. Cardiovascular human organ-on-a-chip platform for disease modeling, drug development, and personalized therapy. J Biomed Mater Res A 2024; 112:512-523. [PMID: 37668192 PMCID: PMC11089005 DOI: 10.1002/jbm.a.37602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/16/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular organ-on-a-chip (OoC) devices are composed of engineered or native functional tissues that are cultured under controlled microenvironments inside microchips. These systems employ microfabrication and tissue engineering techniques to recapitulate human physiology. This review focuses on human OoC systems to model cardiovascular diseases, to perform drug screening, and to advance personalized medicine. We also address the challenges in the generation of organ chips that can revolutionize the large-scale application of these systems for drug development and personalized therapy.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
8
|
Shu Q, Wang Y, Lin X, Xie S, Wang Z, Wang S, Yin L. Assessment of fetal intraventricular diastolic fluid dynamics using ultrasound vector flow mapping. BMC Cardiovasc Disord 2023; 23:488. [PMID: 37794371 PMCID: PMC10552239 DOI: 10.1186/s12872-023-03524-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
OBJECTIVE The purpose of this study was to investigate the feasibility of visualizing and quantifying the normal pattern of vortex formation in the left ventricle (LV) and right ventricle (RV) of the fetal heart during diastole using vector flow mapping (VFM). METHODS A total of 36 healthy fetuses in the second trimester (mean gestational age: 23 weeks, 2 days; range: 22-24 weeks) were enrolled in the study. Color Doppler signals were recorded in the four-chamber view to observe the phase of the diastolic vortices in the LV and RV. The vortex area and circulation were measured, and parameters such as intraventricular pressure difference (IVPD), intraventricular pressure gradient (IVPG), and average energy loss (EL_AVG) were evaluated at different diastolic phases, including isovolumic relaxation (D1), early diastole (D2), and late diastole (D3). RESULTS Healthy second-trimester fetal vortex formations were observed in both the LV and RV at the end of diastole, with the vortices rotating in a clockwise direction towards the outflow tract. There were no significant differences in vortex area and circulation between the two ventricles (p > 0.05). However, significant differences were found in IVPD, IVPG, and EL_AVG among the diastolic phases (D1, D2, and D3) (p < 0.05). Trends in IVPD, IVPG, and EL_AVG during diastole (D1-D2-D3) revealed increasing IVPD and EL_AVG values, as well as decreasing IVPG values. Furthermore, during D3, the RV exhibited significantly higher IVPD, IVPG, and EL_AVG compared to the LV (p > 0.05). CONCLUSION VFM is a valuable technique for analyzing the formation of vortices in the left and right ventricles during fetal diastole. The application of VFM technology has the potential to enhance the assessment of fetal cardiac parameters.
Collapse
Affiliation(s)
- Qinglan Shu
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyi Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shenghua Xie
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhengyang Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Sijia Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lixue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Department of Cardiovascular Ultrasound & Noninvasive Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
9
|
Dou W, Daoud A, Chen X, Wang T, Malhi M, Gong Z, Mirshafiei F, Zhu M, Shan G, Huang X, Maynes JT, Sun Y. Ultrathin and Flexible Bioelectronic Arrays for Functional Measurement of iPSC-Cardiomyocytes under Cardiotropic Drug Administration and Controlled Microenvironments. NANO LETTERS 2023; 23:2321-2331. [PMID: 36893018 DOI: 10.1021/acs.nanolett.3c00017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Emerging heart-on-a-chip technology is a promising tool to establish in vitro cardiac models for therapeutic testing and disease modeling. However, due to the technical complexity of integrating cell culture chambers, biosensors, and bioreactors into a single entity, a microphysiological system capable of reproducing controlled microenvironmental cues to regulate cell phenotypes, promote iPS-cardiomyocyte maturity, and simultaneously measure the dynamic changes of cardiomyocyte function in situ is not available. This paper reports an ultrathin and flexible bioelectronic array platform in 24-well format for higher-throughput contractility measurement under candidate drug administration or defined microenvironmental conditions. In the array, carbon black (CB)-PDMS flexible strain sensors were embedded for detecting iPSC-CM contractility signals. Carbon fiber electrodes and pneumatic air channels were integrated to provide electrical and mechanical stimulation to improve iPSC-CM maturation. Performed experiments validate that the bioelectronic array accurately reveals the effects of cardiotropic drugs and identifies mechanical/electrical stimulation strategies for promoting iPSC-CM maturation.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Abdelkader Daoud
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Tiancong Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Zheyuan Gong
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Fatemeh Mirshafiei
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Min Zhu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Jason T Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario M5S 3G4, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario M5T 3A1, Canada
| |
Collapse
|
10
|
Gabetti S, Sileo A, Montrone F, Putame G, Audenino AL, Marsano A, Massai D. Versatile electrical stimulator for cardiac tissue engineering-Investigation of charge-balanced monophasic and biphasic electrical stimulations. Front Bioeng Biotechnol 2023; 10:1031183. [PMID: 36686253 PMCID: PMC9846083 DOI: 10.3389/fbioe.2022.1031183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023] Open
Abstract
The application of biomimetic physical stimuli replicating the in vivo dynamic microenvironment is crucial for the in vitro development of functional cardiac tissues. In particular, pulsed electrical stimulation (ES) has been shown to improve the functional properties of in vitro cultured cardiomyocytes. However, commercially available electrical stimulators are expensive and cumbersome devices while customized solutions often allow limited parameter tunability, constraining the investigation of different ES protocols. The goal of this study was to develop a versatile compact electrical stimulator (ELETTRA) for biomimetic cardiac tissue engineering approaches, designed for delivering controlled parallelizable ES at a competitive cost. ELETTRA is based on an open-source micro-controller running custom software and is combinable with different cell/tissue culture set-ups, allowing simultaneously testing different ES patterns on multiple samples. In particular, customized culture chambers were appositely designed and manufactured for investigating the influence of monophasic and biphasic pulsed ES on cardiac cell monolayers. Finite element analysis was performed for characterizing the spatial distributions of the electrical field and the current density within the culture chamber. Performance tests confirmed the accuracy, compliance, and reliability of the ES parameters delivered by ELETTRA. Biological tests were performed on neonatal rat cardiac cells, electrically stimulated for 4 days, by comparing, for the first time, the monophasic waveform (electric field = 5 V/cm) to biphasic waveforms by matching either the absolute value of the electric field variation (biphasic ES at ±2.5 V/cm) or the total delivered charge (biphasic ES at ±5 V/cm). Findings suggested that monophasic ES at 5 V/cm and, particularly, charge-balanced biphasic ES at ±5 V/cm were effective in enhancing electrical functionality of stimulated cardiac cells and in promoting synchronous contraction.
Collapse
Affiliation(s)
- Stefano Gabetti
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Antonio Sileo
- Department of Surgery and Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Federica Montrone
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Giovanni Putame
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Alberto L. Audenino
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Anna Marsano
- Department of Surgery and Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Diana Massai
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy,*Correspondence: Diana Massai,
| |
Collapse
|
11
|
A maladaptive feedback mechanism between the extracellular matrix and cytoskeleton contributes to hypertrophic cardiomyopathy pathophysiology. Commun Biol 2023; 6:4. [PMID: 36596888 PMCID: PMC9810744 DOI: 10.1038/s42003-022-04278-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 11/17/2022] [Indexed: 01/04/2023] Open
Abstract
Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.
Collapse
|
12
|
Karamali F, Behtaj S, Babaei-Abraki S, Hadady H, Atefi A, Savoj S, Soroushzadeh S, Najafian S, Nasr Esfahani MH, Klassen H. Potential therapeutic strategies for photoreceptor degeneration: the path to restore vision. J Transl Med 2022; 20:572. [PMID: 36476500 PMCID: PMC9727916 DOI: 10.1186/s12967-022-03738-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 12/12/2022] Open
Abstract
Photoreceptors (PRs), as the most abundant and light-sensing cells of the neuroretina, are responsible for converting light into electrical signals that can be interpreted by the brain. PR degeneration, including morphological and functional impairment of these cells, causes significant diminution of the retina's ability to detect light, with consequent loss of vision. Recent findings in ocular regenerative medicine have opened promising avenues to apply neuroprotective therapy, gene therapy, cell replacement therapy, and visual prostheses to the challenge of restoring vision. However, successful visual restoration in the clinical setting requires application of these therapeutic approaches at the appropriate stage of the retinal degeneration. In this review, firstly, we discuss the mechanisms of PR degeneration by focusing on the molecular mechanisms underlying cell death. Subsequently, innovations, recent developments, and promising treatments based on the stage of disorder progression are further explored. Then, the challenges to be addressed before implementation of these therapies in clinical practice are considered. Finally, potential solutions to overcome the current limitations of this growing research area are suggested. Overall, the majority of current treatment modalities are still at an early stage of development and require extensive additional studies, both pre-clinical and clinical, before full restoration of visual function in PR degeneration diseases can be realized.
Collapse
Affiliation(s)
- Fereshteh Karamali
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sanaz Behtaj
- grid.1022.10000 0004 0437 5432Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia ,grid.1022.10000 0004 0437 5432Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222 Australia
| | - Shahnaz Babaei-Abraki
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Hadady
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Soraya Savoj
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sareh Soroushzadeh
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Henry Klassen
- grid.266093.80000 0001 0668 7243Gavin Herbert Eye Institute, Irvine, CA USA
| |
Collapse
|
13
|
Extracellular stiffness induces contractile dysfunction in adult cardiomyocytes via cell-autonomous and microtubule-dependent mechanisms. Basic Res Cardiol 2022; 117:41. [PMID: 36006489 PMCID: PMC9899517 DOI: 10.1007/s00395-022-00952-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023]
Abstract
The mechanical environment of the myocardium has a potent effect on cardiomyocyte form and function, yet an understanding of the cardiomyocyte responses to extracellular stiffening remains incomplete. We therefore employed a cell culture substrate with tunable stiffness to define the cardiomyocyte responses to clinically relevant stiffness increments in the absence of cell-cell interactions. When cultured on substrates magnetically actuated to mimic the stiffness of diseased myocardium, isolated rat adult cardiomyocytes exhibited a time-dependent reduction of sarcomere shortening, characterized by slowed contraction and relaxation velocity, and alterations of the calcium transient. Cardiomyocytes cultured on stiff substrates developed increases in viscoelasticity and microtubule detyrosination in association with early increases in the α-tubulin detyrosinating enzyme vasohibin-2 (Vash2). We found that knockdown of Vash2 was sufficient to preserve contractile performance as well as calcium transient properties in the presence of extracellular substrate stiffening. Orthogonal prevention of detyrosination by overexpression of tubulin tyrosine ligase (TTL) was also able to preserve contractility and calcium homeostasis. These data demonstrate that a pathologic increment of extracellular stiffness induces early, cell-autonomous remodeling of adult cardiomyocytes that is dependent on detyrosination of α-tubulin.
Collapse
|
14
|
Strimaityte D, Tu C, Yanez A, Itzhaki I, Wu H, Wu JC, Yang H. Contractility and Calcium Transient Maturation in the Human iPSC-Derived Cardiac Microfibers. ACS APPLIED MATERIALS & INTERFACES 2022; 14:35376-35388. [PMID: 35901275 PMCID: PMC9780031 DOI: 10.1021/acsami.2c07326] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are considered immature in the sarcomere organization, contractile machinery, calcium transient, and transcriptome profile, which prevent them from further applications in modeling and studying cardiac development and disease. To improve the maturity of hiPSC-CMs, here, we engineered the hiPSC-CMs into cardiac microfibers (iCMFs) by a stencil-based micropatterning method, which enables the hiPSC-CMs to be aligned in an end-to-end connection for prolonged culture on the hydrogel of physiological stiffness. A series of characterization approaches were performed to evaluate the maturation in iCMFs on both structural and functional levels, including immunohistochemistry, calcium transient, reverse-transcription quantitative PCR, cardiac contractility, and electrical pacing analysis. Our results demonstrate an improved cardiac maturation of hiPSC-CMs in iCMFs compared to micropatterned or random single hiPSC-CMs and hiPSC-CMs in a random cluster at the same cell number of iCMFs. We found an increased sarcomere length, better regularity and alignment of sarcomeres, enhanced contractility, matured calcium transient, and T-tubule formation and improved adherens junction and gap junction formation. The hiPSC-CMs in iCMFs showed a robust calcium cycling in response to the programmed and continuous electrical pacing from 0.5 to 7 Hz. Moreover, we generated the iCMFs with hiPSC-CMs with mutations in myosin-binding protein C (MYBPC3) to have a proof-of-concept of iCMFs in modeling cardiac hypertrophic phenotype. These findings suggest that the multipatterned iCMF connection of hiPSC-CMs boosts the cardiac maturation structurally and functionally, which will reveal the full potential of the application of hiPSC-CM models in disease modeling of cardiomyopathy and cardiac regenerative medicine.
Collapse
Affiliation(s)
- Dovile Strimaityte
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA
| | - Chengyi Tu
- Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Apuleyo Yanez
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA
| | - Ilanit Itzhaki
- Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Haodi Wu
- Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Joseph C. Wu
- Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA
| |
Collapse
|
15
|
Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Suji Choi
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
| | - Christina Alamana
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kevin K. Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
16
|
Rashid SA, Blanchard AT, Combs JD, Fernandez N, Dong Y, Cho HC, Salaita K. DNA Tension Probes Show that Cardiomyocyte Maturation Is Sensitive to the Piconewton Traction Forces Transmitted by Integrins. ACS NANO 2022; 16:5335-5348. [PMID: 35324164 PMCID: PMC11238821 DOI: 10.1021/acsnano.1c04303] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cardiac muscle cells (CMCs) are the unit cells that comprise the heart. CMCs go through different stages of differentiation and maturation pathways to fully mature into beating cells. These cells can sense and respond to mechanical cues through receptors such as integrins which influence maturation pathways. For example, cell traction forces are important for the differentiation and development of functional CMCs, as CMCs cultured on varying substrate stiffness function differently. Most work in this area has focused on understanding the role of bulk extracellular matrix stiffness in mediating the functional fate of CMCs. Given that stiffness sensing mechanisms are mediated by individual integrin receptors, an important question in this area pertains to the specific magnitude of integrin piconewton (pN) forces that can trigger CMC functional maturation. To address this knowledge gap, we used DNA adhesion tethers that rupture at specific thresholds of force (∼12, ∼56, and ∼160 pN) to test whether capping peak integrin tension to specific magnitudes affects CMC function. We show that adhesion tethers with greater force tolerance lead to functionally mature CMCs as determined by morphology, twitching frequency, transient calcium flux measurements, and protein expression (F-actin, vinculin, α-actinin, YAP, and SERCA2a). Additionally, sarcomeric actinin alignment and multinucleation were significantly enhanced as the mechanical tolerance of integrin tethers was increased. Taken together, the results show that CMCs harness defined pN integrin forces to influence early stage development. This study represents an important step toward biophysical characterization of the contribution of pN forces in early stage cardiac differentiation.
Collapse
Affiliation(s)
- Sk Aysha Rashid
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Aaron T Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - J Dale Combs
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Natasha Fernandez
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 1405 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Hee Cheol Cho
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 1405 Clifton Road NE, Atlanta, Georgia 30322, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Khalid Salaita
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| |
Collapse
|
17
|
Fang Y, Fan W, Xu X, Janoshazi AK, Fargo DC, Li X. SIRT1 regulates cardiomyocyte alignment during maturation. J Cell Sci 2022; 135:274667. [PMID: 35260907 PMCID: PMC9016619 DOI: 10.1242/jcs.259076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocyte elongation and alignment, a critical step in cardiomyocyte maturation starting from the perinatal stage, is crucial for formation of the highly organized intra- and inter-cellular structures for spatially and temporally ordered contraction in adult cardiomyocytes. However, the mechanism(s) underlying the control of cardiomyocyte alignment remains elusive. Here, we report that SIRT1, the most conserved NAD+-dependent protein deacetylase highly expressed in perinatal heart, plays an important role in regulating cardiomyocyte remodeling during development. We observed that SIRT1 deficiency impairs the alignment of cardiomyocytes/myofibrils and disrupts normal beating patterns at late developmental stages in an in vitro differentiation system from human embryonic stem cells. Consistently, deletion of SIRT1 at a late developmental stage in mouse embryos induced the irregular distribution of cardiomyocytes and misalignment of myofibrils, and reduced the heart size. Mechanistically, the expression of several genes involved in chemotaxis, including those in the CXCL12/CXCR4 and CCL2/CCR2/CCR4 pathways, was dramatically blunted during maturation of SIRT1-deficient cardiomyocytes. Pharmacological inhibition of CCL2 signaling suppressed cardiomyocyte alignment. Our study identifies a regulatory factor that modulates cardiomyocyte alignment at the inter-cellular level during maturation.
Collapse
Affiliation(s)
- Yi Fang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA,Authors for correspondence (; )
| | - Wei Fan
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Agnes K. Janoshazi
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA,Fluorescence Microscopy and Imaging Center, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David C. Fargo
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA,Authors for correspondence (; )
| |
Collapse
|
18
|
Zevolis E, Philippou A, Moustogiannis A, Chatzigeorgiou A, Koutsilieris M. The Effects of Mechanical Loading Variations on the Hypertrophic, Anti-Apoptotic, and Anti-Inflammatory Responses of Differentiated Cardiomyocyte-like H9C2 Cells. Cells 2022; 11:473. [PMID: 35159283 PMCID: PMC8834179 DOI: 10.3390/cells11030473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/25/2021] [Accepted: 01/26/2022] [Indexed: 11/28/2022] Open
Abstract
Cardiomyocytes possess the ability to respond to mechanical stimuli by adapting their biological functions. This study investigated cellular and molecular events in cardiomyocyte-like H9C2 cells during differentiation as well as the signalling and gene expression responses of the differentiated cells under various mechanical stretching protocols in vitro. Immunofluorescence was used to monitor MyHC expression and structural changes during cardiomyoblast differentiation. Moreover, alterations in the expression of cardiac-specific markers, cell cycle regulatory factors, MRFs, hypertrophic, apoptotic, atrophy and inflammatory factors, as well as the activation of major intracellular signalling pathways were evaluated during differentiation and under mechanical stretching of the differentiated H9C2 cells. Compared to undifferentiated cells, advanced-differentiation cardiomyoblasts exhibited increased expression of cardiac-specific markers, MyHC, MRFs, and IGF-1 isoforms. Moreover, differentiated cells that underwent a low strain/frequency mechanical loading protocol of intermediate duration showed enhanced expression of MRFs and hypertrophic factors, along with a decreased expression of apoptotic, atrophy, and inflammatory factors compared to both high-strain/frequency loading protocols and to unloaded cells. These findings suggest that altering the strain and frequency of mechanical loading applied on differentiated H9C2 cardiomyoblasts can regulate their anabolic/survival program, with a low-strain/frequency stretching being, overall, most effective at inducing beneficial responses.
Collapse
Affiliation(s)
| | | | | | | | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27 Athens, Greece; (E.Z.); (A.P.); (A.M.); (A.C.)
| |
Collapse
|
19
|
Alvarez-Dominguez JR, Melton DA. Cell maturation: Hallmarks, triggers, and manipulation. Cell 2022; 185:235-249. [PMID: 34995481 PMCID: PMC8792364 DOI: 10.1016/j.cell.2021.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
How cells become specialized, or "mature," is important for cell and developmental biology. While maturity is usually deemed a terminal fate, it may be more helpful to consider maturation not as a switch but as a dynamic continuum of adaptive phenotypic states set by genetic and environment programing. The hallmarks of maturity comprise changes in anatomy (form, gene circuitry, and interconnectivity) and physiology (function, rhythms, and proliferation) that confer adaptive behavior. We discuss efforts to harness their chemical (nutrients, oxygen, and growth factors) and physical (mechanical, spatial, and electrical) triggers in vitro and in vivo and how maturation strategies may support disease research and regenerative medicine.
Collapse
Affiliation(s)
- Juan R. Alvarez-Dominguez
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Douglas A. Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
20
|
Liu J, Lian H, Yu J, Wu J, Chen X, wang P, tian L, Yang Y, Yang J, Li D, Guo S. Study on diverse pathological characteristics of heart failure in different stages based on proteomics. J Cell Mol Med 2022; 26:1169-1182. [PMID: 35048506 PMCID: PMC8831959 DOI: 10.1111/jcmm.17170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 11/30/2022] Open
Abstract
Heart failure is a process characterized by significant disturbance of protein turnover. To elucidate the alterations in cardiac protein expression during the various phases of heart failure and to understand the nature of the processes involved, we analysed the proteome in an established heart failure model at different time points to monitor thousands of different proteins simultaneously. Here, heart failure was induced by transverse aortic constriction (TAC) in KM mice. At 2, 4 and 12 weeks after operation, protein expression profiles were determined in sham‐operated (controls) and TAC mice, using label‐free quantitative proteomics, leading to identification and quantification of almost 4000 proteins. The results of the KEGG pathway enrichment analysis and GO function annotation revealed critical pathways associated with the transition from cardiac hypertrophy to heart failure, such as energy pathways and matrix reorganization. Our study suggests that in the pathophysiology of heart failure, alterations of protein groups related to cardiac energy substrate metabolism and cytoskeleton remodelling could play the more dominant roles for the signalling that eventually results in contractile dysfunction and heart failure.
Collapse
Affiliation(s)
- Jinying Liu
- College of Traditional Chinese Medicine Chengde Medical University Chengde Hebei Province China
- School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Hongjian Lian
- School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
- Alexa League Central Hospital Inner Mongolia China
| | - Jiang Yu
- School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Jie Wu
- College of Traditional Chinese Medicine Chengde Medical University Chengde Hebei Province China
| | - Xiangyang Chen
- Youcare Pharmaceutical Group Drug Research Institute Beijing China
| | - Peng wang
- College of Traditional Chinese Medicine Chengde Medical University Chengde Hebei Province China
| | - Lei tian
- School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Yunfei Yang
- Beijing Qinglian Biotech Co., Ltd Beijing China
| | - Jiaqi Yang
- College of Traditional Chinese Medicine Chengde Medical University Chengde Hebei Province China
| | - Dong Li
- School of Basic Medical Sciences Anhui Medical University Hefei China
- State Key Laboratory of Proteomics Beijing Proteome Research Center National Center for Protein Sciences (PHOENIX Center) Beijing Institute of Lifeomics Beijing China
| | - Shuzhen Guo
- School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| |
Collapse
|
21
|
Myocardial Afterload Is a Key Biomechanical Regulator of Atrioventricular Myocyte Differentiation in Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010022. [PMID: 35050232 PMCID: PMC8779957 DOI: 10.3390/jcdd9010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Heart valve development is governed by both genetic and biomechanical inputs. Prior work has demonstrated that oscillating shear stress associated with blood flow is required for normal atrioventricular (AV) valve development. Cardiac afterload is defined as the pressure the ventricle must overcome in order to pump blood throughout the circulatory system. In human patients, conditions of high afterload can cause valve pathology. Whether high afterload adversely affects embryonic valve development remains poorly understood. Here we describe a zebrafish model exhibiting increased myocardial afterload, caused by vasopressin, a vasoconstrictive drug. We show that the application of vasopressin reliably produces an increase in afterload without directly acting on cardiac tissue in zebrafish embryos. We have found that increased afterload alters the rate of growth of the cardiac chambers and causes remodeling of cardiomyocytes. Consistent with pathology seen in patients with clinically high afterload, we see defects in both the form and the function of the valve leaflets. Our results suggest that valve defects are due to changes in atrioventricular myocyte signaling, rather than pressure directly acting on the endothelial valve leaflet cells. Cardiac afterload should therefore be considered a biomechanical factor that particularly impacts embryonic valve development.
Collapse
|
22
|
Müller D, Donath S, Brückner EG, Biswanath Devadas S, Daniel F, Gentemann L, Zweigerdt R, Heisterkamp A, Kalies SMK. How Localized Z-Disc Damage Affects Force Generation and Gene Expression in Cardiomyocytes. Bioengineering (Basel) 2021; 8:bioengineering8120213. [PMID: 34940366 PMCID: PMC8698600 DOI: 10.3390/bioengineering8120213] [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: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022] Open
Abstract
The proper function of cardiomyocytes (CMs) is highly related to the Z-disc, which has a pivotal role in orchestrating the sarcomeric cytoskeletal function. To better understand Z-disc related cardiomyopathies, novel models of Z-disc damage have to be developed. Human pluripotent stem cell (hPSC)-derived CMs can serve as an in vitro model to better understand the sarcomeric cytoskeleton. A femtosecond laser system can be applied for localized and defined damage application within cells as single Z-discs can be removed. We have investigated the changes in force generation via traction force microscopy, and in gene expression after Z-disc manipulation in hPSC-derived CMs. We observed a significant weakening of force generation after removal of a Z-disc. However, no significant changes of the number of contractions after manipulation were detected. The stress related gene NF-kB was significantly upregulated. Additionally, α-actinin (ACTN2) and filamin-C (FLNc) were upregulated, pointing to remodeling of the Z-disc and the sarcomeric cytoskeleton. Ultimately, cardiac troponin I (TNNI3) and cardiac muscle troponin T (TNNT2) were significantly downregulated. Our results allow a better understanding of transcriptional coupling of Z-disc damage and the relation of damage to force generation and can therefore finally pave the way to novel therapies of sarcomeric disorders.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Sören Donath
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Emanuel Georg Brückner
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Santoshi Biswanath Devadas
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Fiene Daniel
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Robert Zweigerdt
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Stefan Michael Klaus Kalies
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
23
|
Roth BJ. Bidomain modeling of electrical and mechanical properties of cardiac tissue. BIOPHYSICS REVIEWS 2021; 2:041301. [PMID: 38504719 PMCID: PMC10903405 DOI: 10.1063/5.0059358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/15/2021] [Indexed: 03/21/2024]
Abstract
Throughout the history of cardiac research, there has been a clear need to establish mathematical models to complement experimental studies. In an effort to create a more complete picture of cardiac phenomena, the bidomain model was established in the late 1970s to better understand pacing and defibrillation in the heart. This mathematical model has seen ongoing use in cardiac research, offering mechanistic insight that could not be obtained from experimental pursuits. Introduced from a historical perspective, the origins of the bidomain model are reviewed to provide a foundation for researchers new to the field and those conducting interdisciplinary research. The interplay of theory and experiment with the bidomain model is explored, and the contributions of this model to cardiac biophysics are critically evaluated. Also discussed is the mechanical bidomain model, which is employed to describe mechanotransduction. Current challenges and outstanding questions in the use of the bidomain model are addressed to give a forward-facing perspective of the model in future studies.
Collapse
Affiliation(s)
- Bradley J. Roth
- Department of Physics, Oakland University, Rochester, Michigan 48309, USA
| |
Collapse
|
24
|
Qin Y, Hu X, Fan W, Yan J, Cheng S, Liu Y, Huang W. A Stretchable Scaffold with Electrochemical Sensing for 3D Culture, Mechanical Loading, and Real-Time Monitoring of Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003738. [PMID: 34047055 PMCID: PMC8327466 DOI: 10.1002/advs.202003738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/24/2020] [Indexed: 06/11/2023]
Abstract
In the field of three-dimensional (3D) cell culture and tissue engineering, great advance focusing on functionalized materials and desirable culture systems has been made to mimic the natural environment of cells in vivo. Mechanical loading is one of the critical factors that affect cell/tissue behaviors and metabolic activities, but the reported models or detection methods offer little direct and real-time information about mechanically induced cell responses. Herein, for the first time, a stretchable and multifunctional platform integrating 3D cell culture, mechanical loading, and electrochemical sensing is developed by immobilization of biomimetic peptide linked gold nanotubes on porous and elastic polydimethylsiloxane. The 3D scaffold demonstrates very good compatibility, excellent stretchability, and stable electrochemical sensing performance. This allows mimicking the articular cartilage and investigating its mechanotransduction by 3D culture, mechanical stretching of chondrocytes, and synchronously real-time monitoring of stretch-induced signaling molecules. The results disclose a previously unclear mechanotransduction pathway in chondrocytes that mechanical loading can rapidly activate nitric oxide signaling within seconds. This indicates the promising potential of the stretchable 3D sensing in exploring the mechanotransduction in 3D cellular systems and engineered tissues.
Collapse
Affiliation(s)
- Yu Qin
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| | - Xue‐Bo Hu
- College of Chemistry and Chemical EngineeringInstitute for Conservation and Utilization of Agro‐Bioresources in Dabie MountainsXinyang Normal UniversityXinyang464000China
| | - Wen‐Ting Fan
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| | - Jing Yan
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| | - Shi‐Bo Cheng
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| | - Yan‐Ling Liu
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| | - Wei‐Hua Huang
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072China
| |
Collapse
|
25
|
Iskander A, Bilgi C, Naftalovich R, Hacihaliloglu I, Berkman T, Naftalovich D, Pahlevan N. The Rheology of the Carotid Sinus: A Path Toward Bioinspired Intervention. Front Bioeng Biotechnol 2021; 9:678048. [PMID: 34178967 PMCID: PMC8222608 DOI: 10.3389/fbioe.2021.678048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022] Open
Abstract
The association between blood viscosity and pathological conditions involving a number of organ systems is well known. However, how the body measures and maintains appropriate blood viscosity is not well-described. The literature endorsing the function of the carotid sinus as a site of baroreception can be traced back to some of the earliest descriptions of digital pressure on the neck producing a drop in blood delivery to the brain. For the last 30 years, improved computational fluid dynamic (CFD) simulations of blood flow within the carotid sinus have demonstrated a more nuanced understanding of the changes in the region as it relates to changes in conventional metrics of cardiovascular function, including blood pressure. We suggest that the unique flow patterns within the carotid sinus may make it an ideal site to transduce flow data that can, in turn, enable real-time measurement of blood viscosity. The recent characterization of the PIEZO receptor family in the sinus vessel wall may provide a biological basis for this characterization. When coupled with other biomarkers of cardiovascular performance and descriptions of the blood rheology unique to the sinus region, this represents a novel venue for bioinspired design that may enable end-users to manipulate and optimize blood flow.
Collapse
Affiliation(s)
- Andrew Iskander
- Department of Anesthesiology, Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Coskun Bilgi
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Rotem Naftalovich
- Department of Anesthesiology, New Jersey Medical School, University Hospital, Rutgers University, Newark, NJ, United States.,Medical Corps of the U.S. Army, U.S. Army Medical Department, Fort Sam Houston, San Antonio, TX, United States
| | - Ilker Hacihaliloglu
- Department of Biomedical Engineering, Rutgers School of Engineering, Rutgers University, Piscataway, NJ, United States
| | - Tolga Berkman
- Department of Anesthesiology, New Jersey Medical School, University Hospital, Rutgers University, Newark, NJ, United States
| | - Daniel Naftalovich
- Department of Computational and Mathematical Sciences, California Institute of Technology, Pasadena, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Niema Pahlevan
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
26
|
Lang J, Li Y, Ye Z, Yang Y, Xu F, Huang G, Zhang J, Li F. Investigating the Effect of Substrate Stiffness on the Redox State of Cardiac Fibroblasts Using Scanning Electrochemical Microscopy. Anal Chem 2021; 93:5797-5804. [PMID: 33797232 DOI: 10.1021/acs.analchem.0c05284] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cardiac fibrosis, in which cardiac fibroblasts differentiate into myofibroblasts, leads to oversecretion of the extracellular matrix, results in increased stiffness, and facilitates disequilibrium of cellular redox state, further leading to oxidative stress and various degrees of cell death. However, the relationship between the matrix stiffness and the redox status of cardiac fibroblasts remains unclear. In this work, we constructed an in vitro cardiac fibrosis model by culturing cardiac fibroblasts on polyacrylamide gels with tunable stiffness and characterized the differentiation of cardiac fibroblasts to myofibroblasts by immunofluorescence staining of α-smooth muscle actin. We then applied scanning electrochemical microscopy (SECM) with a depth scan mode to in situ and quantitatively assess the redox status by monitoring the glutathione (GSH) efflux rate (k) through the redox reaction between GSH (a typical indicator of cellular redox level) released from cardiac fibroblasts and SECM probe-oxidized ferrocenecarboxylic acid ([FcCOOH]+). The SECM results demonstrate that the GSH efflux from the cardiac fibroblasts decreased with increasing substrate stiffness (i.e., mimicking the increased fibrosis degree), indicating that a more oxidizing microenvironment facilitates the cell differentiation and GSH may serve as a biomarker to predict the degree of cardiac fibrosis. This work provides an SECM approach to quantify the redox state of cardiac fibroblasts by recording the GSH efflux rate. In addition, the newly established relationship between the redox balance and the substrate stiffness would help to better understand the redox state of cardiac fibroblasts during cardiac fibrosis.
Collapse
Affiliation(s)
- Jinxin Lang
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yabei Li
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhaoyang Ye
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yaowei Yang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan 430072, P. R. China
| | - Junjie Zhang
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
27
|
Lui C, Chin AF, Park S, Yeung E, Kwon C, Tomaselli G, Chen Y, Hibino N. Mechanical stimulation enhances development of scaffold-free, 3D-printed, engineered heart tissue grafts. J Tissue Eng Regen Med 2021; 15:503-512. [PMID: 33749089 DOI: 10.1002/term.3188] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/10/2021] [Indexed: 12/26/2022]
Abstract
Current efforts to engineer a clinically relevant tissue graft from human-induced pluripotent stem cells (hiPSCs) have relied on the addition or utilization of external scaffolding material. However, any imbalance in the interactions between embedded cells and their surroundings may hinder the success of the resulting tissue graft. Therefore, the goal of our study was to create scaffold-free, 3D-printed cardiac tissue grafts from hiPSC-derived cardiomyocytes (CMs), and to evaluate whether or not mechanical stimulation would result in improved graft maturation. To explore this, we used a 3D bioprinter to produce scaffold-free cardiac tissue grafts from hiPSC-derived CM cell spheroids. Static mechanical stretching of these grafts significantly increased sarcomere length compared to unstimulated free-floating tissues, as determined by immunofluorescent image analysis. Stretched tissue was found to have decreased elastic modulus, increased maximal contractile force, and increased alignment of formed extracellular matrix, as expected in a functionally maturing tissue graft. Additionally, stretched tissues had upregulated expression of cardiac-specific gene transcripts, consistent with increased cardiac-like cellular identity. Finally, analysis of extracellular matrix organization in stretched grafts suggests improved remodeling by embedded cardiac fibroblasts. Taken together, our results suggest that mechanical stretching stimulates hiPSC-derived CMs in a 3D-printed, scaffold-free tissue graft to develop mature cardiac material structuring and cellular fates. Our work highlights the critical role of mechanical conditioning as an important engineering strategy toward developing clinically applicable, scaffold-free human cardiac tissue grafts.
Collapse
Affiliation(s)
- Cecillia Lui
- Division of Cardiac Surgery, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Alexander F Chin
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Enoch Yeung
- Division of Cardiac Surgery, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Chulan Kwon
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gordon Tomaselli
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Narutoshi Hibino
- Division of Cardiac Surgery, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Liu T, Zhang S, Huang C, Ma S, Bai R, Li Y, Chang Y, Hang C, Saleem A, Dong T, Guo T, Jiang Y, Lu W, Zhang L, Jianwen L, Jiang H, Lan F. Microscale grooves regulate maturation development of hPSC-CMs by the transient receptor potential channels (TRP channels). J Cell Mol Med 2021; 25:3469-3483. [PMID: 33689230 PMCID: PMC8034460 DOI: 10.1111/jcmm.16429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The use of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is limited in drug discovery and cardiac disease mechanism studies due to cell immaturity. Micro-scaled grooves can promote the maturation of cardiomyocytes by aligning them in order, but the mechanism of cardiomyocytes alignment has not been studied. From the level of calcium activity, gene expression and cell morphology, we verified that the W20H5 grooves can effectively promote the maturation of cardiomyocytes. The transient receptor potential channels (TRP channels) also play an important role in the maturation and development of cardiomyocytes. These findings support the engineered hPSC-CMs as a powerful model to study cardiac disease mechanism and partly mimic the myocardial morphological development. The important role of the TRP channels in the maturation and development of myocardium is first revealed.
Collapse
Affiliation(s)
- Taoyan Liu
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Siyao Zhang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Chenwu Huang
- Department of Biomedical EngineeringSchool of MedicineTsinghua UniversityBeijingChina
| | - Shuhong Ma
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Rui Bai
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Yanan Li
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Yun Chang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Chenwen Hang
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Amina Saleem
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Tao Dong
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Tianwei Guo
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Youxu Jiang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Wenjing Lu
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Lina Zhang
- State Key Laboratory of Chemical Resource EngineeringBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Luo Jianwen
- Department of Biomedical EngineeringSchool of MedicineTsinghua UniversityBeijingChina
| | - Hongfeng Jiang
- Key Laboratory of Remodeling‐Related Cardiovascular DiseasesMinistry of EducationBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Feng Lan
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
29
|
Öztürk-Öncel MÖ, Heras-Bautista CO, Uzun L, Hür D, Hescheler J, Pfannkuche K, Garipcan B. Impact of Poly(dimethylsiloxane) Surface Modification with Conventional and Amino Acid-Conjugated Self-Assembled Monolayers on the Differentiation of Induced Pluripotent Stem Cells into Cardiomyocytes. ACS Biomater Sci Eng 2021; 7:1539-1551. [DOI: 10.1021/acsbiomaterials.0c01434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- M. Özgen Öztürk-Öncel
- Institute of Biomedical Engineering, Boğaziçi University, Kandilli Campus, Rasathane Cd. Uskudar, 34684 Istanbul, Turkey
| | - Carlos O. Heras-Bautista
- Center of Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Lokman Uzun
- Department of Chemistry, Hacettepe University, Beytepe Campus, 06800 Ankara, Turkey
| | - Deniz Hür
- Department of Chemistry, Eskisehir Technical University, 2 Eylül Campus Tepebaşı, 26555 Eskisehir, Turkey
| | - Jürgen Hescheler
- Center of Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Kurt Pfannkuche
- Center of Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Bora Garipcan
- Institute of Biomedical Engineering, Boğaziçi University, Kandilli Campus, Rasathane Cd. Uskudar, 34684 Istanbul, Turkey
| |
Collapse
|
30
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
31
|
Yu J, Cai P, Chen X. Structural Regulation of Myocytes in Engineered Healthy and Diseased Cardiac Models. ACS APPLIED BIO MATERIALS 2021; 4:267-276. [DOI: 10.1021/acsabm.0c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Yu
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Pingqiang Cai
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Xiaodong Chen
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
32
|
Lyra-Leite DM, Petersen AP, Ariyasinghe NR, Cho N, McCain ML. Mitochondrial architecture in cardiac myocytes depends on cell shape and matrix rigidity. J Mol Cell Cardiol 2021; 150:32-43. [PMID: 33038389 PMCID: PMC11956898 DOI: 10.1016/j.yjmcc.2020.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/30/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
Contraction of cardiac myocytes depends on energy generated by the mitochondria. During cardiac development and disease, the structure and function of the mitochondrial network in cardiac myocytes is known to remodel in concert with many other factors, including changes in nutrient availability, hemodynamic load, extracellular matrix (ECM) rigidity, cell shape, and maturation of other intracellular structures. However, the independent role of each of these factors on mitochondrial network architecture is poorly understood. In this study, we tested the hypothesis that cell aspect ratio (AR) and ECM rigidity regulate the architecture of the mitochondrial network in cardiac myocytes. To do this, we spin-coated glass coverslips with a soft, moderate, or stiff polymer. Next, we microcontact printed cell-sized rectangles of fibronectin with AR matching cardiac myocytes at various developmental or disease states onto the polymer surface. We then cultured neonatal rat ventricular myocytes on the patterned surfaces and used confocal microscopy and image processing techniques to quantify sarcomeric α-actinin volume, nucleus volume, and mitochondrial volume, surface area, and size distribution. On some substrates, α-actinin volume increased with cell AR but was not affected by ECM rigidity. Nucleus volume was mostly uniform across all conditions. In contrast, mitochondrial volume increased with cell AR on all substrates. Furthermore, mitochondrial surface area to volume ratio decreased as AR increased on all substrates. Large mitochondria were also more prevalent in cardiac myocytes with higher AR. For select AR, mitochondria were also smaller as ECM rigidity increased. Collectively, these results suggest that mitochondrial architecture in cardiac myocytes is strongly influenced by cell shape and moderately influenced by ECM rigidity. These data have important implications for understanding the factors that impact metabolic performance during heart development and disease.
Collapse
Affiliation(s)
- Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Center for Pharmacogenomics, Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, United States of America.
| |
Collapse
|
33
|
Mojumder J, Choy J, Leng S, Zhong L, Kassab G, Lee L. Mechanical stimuli for left ventricular growth during pressure overload. EXPERIMENTAL MECHANICS 2021; 61:131-146. [PMID: 33746236 PMCID: PMC7968380 DOI: 10.1007/s11340-020-00643-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/21/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND The mechanical stimulus (i.e. stress or stretch) for growth occurring in the pressure-overloaded left ventricle (LV) is not exactly known. OBJECTIVE To address this issue, we investigate the correlation between local ventricular growth (indexed by local wall thickness) and the local acute changes in mechanical stimuli after aortic banding. METHODS LV geometric data were extracted from 3D echo measurements at baseline and 2 weeks in the aortic banding swine model (n = 4). We developed and calibrated animal-specific finite element (FE) model of LV mechanics against pressure and volume waveforms measured at baseline. After the simulation of the acute effects of pressure-overload, the local changes of maximum, mean and minimum myocardial stretches and stresses in three orthogonal material directions (i.e., fiber, sheet and sheet-normal) over a cardiac cycle were quantified. Correlation between mechanical quantities and the corresponding measured local changes in wall thickness was quantified using the Pearson correlation number (PCN) and Spearman rank correlation number (SCN). RESULTS At 2 weeks after banding, the average septum thickness decreased from 10.6 ± 2.92mm to 9.49 ± 2.02mm, whereas the LV free-wall thickness increased from 8.69 ± 1.64mm to 9.4 ± 1.22mm. The FE results show strong correlation of growth with the changes in maximum fiber stress (PCN = 0.5471, SCN = 0.5111) and changes in the mean sheet-normal stress (PCN= 0.5266, SCN = 0.5256). Myocardial stretches, however, do not have good correlation with growth. CONCLUSION These results suggest that fiber stress is the mechanical stimuli for LV growth in pressure-overload.
Collapse
Affiliation(s)
- J. Mojumder
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - J.S. Choy
- California Medical Innovations Institute, San Diego, CA, USA
| | - S. Leng
- National Heart Centre Singapore, Singapore
| | - L. Zhong
- National Heart Centre Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore
| | - G.S. Kassab
- California Medical Innovations Institute, San Diego, CA, USA
| | - L.C. Lee
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
34
|
Zevolis E, Philippou A, Moustogiannis A, Chatzigeorgiou A, Koutsilieris M. Optimizing mechanical stretching protocols for hypertrophic and anti-apoptotic responses in cardiomyocyte-like H9C2 cells. Mol Biol Rep 2021; 48:645-655. [PMID: 33394230 DOI: 10.1007/s11033-020-06112-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/18/2020] [Indexed: 01/07/2023]
Abstract
Cardiomyocytes possess the ability to respond to mechanical stimuli by reprogramming their gene expression. This study investigated the effects of different loading protocols on signaling and expression responses of myogenic, anabolic, inflammatory, atrophy and pro-apoptotic genes in cardiomyocyte-like H9C2 cells. Differentiated H9C2 cells underwent various stretching protocols by altering their elongation, frequency and duration, utilizing an in vitro cell tension system. The loading-induced expression changes of MyoD, Myogenin, MRF4, IGF-1 isoforms, Atrogin-1, Foxo1, Fuca and IL-6 were measured by Real Time-PCR. The stretching-induced activation of Akt and Erk 1/2 was also evaluated by Western blot analysis. Low strain (2.7% elongation), low frequency (0.25 Hz) and intermediate duration (12 h) stretching protocol was overall the most effective in inducing beneficial responses, i.e., protein synthesis along with the suppression of apoptosis, inflammation and atrophy, in the differentiated cardiomyocytes. These findings demonstrated that varying the characteristics of mechanical loading applied on H9C2 cells in vitro can regulate their anabolic/survival program.
Collapse
Affiliation(s)
- Evangelos Zevolis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Athens, Greece
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Athens, Greece
| | - Athanasios Moustogiannis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Athens, Greece.
| |
Collapse
|
35
|
Chanthra N, Uosaki H. Maturity of Pluripotent Stem Cell-Derived Cardiomyocytes and Future Perspectives for Regenerative Medicine. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Gendernalik A, Zebhi B, Ahuja N, Garrity D, Bark D. In Vivo Pressurization of the Zebrafish Embryonic Heart as a Tool to Characterize Tissue Properties During Development. Ann Biomed Eng 2020; 49:834-845. [PMID: 32959136 DOI: 10.1007/s10439-020-02619-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022]
Abstract
Cardiac morphogenesis requires an intricate orchestration of mechanical stress to sculpt the heart as it transitions from a straight tube to a multichambered adult heart. Mechanical properties are fundamental to this process, involved in a complex interplay with function, morphology, and mechanotransduction. In the current work, we propose a pressurization technique applied to the zebrafish atrium to quantify mechanical properties of the myocardium under passive tension. By further measuring deformation, we obtain a pressure-stretch relationship that is used to identify constitutive models of the zebrafish embryonic cardiac tissue. Two-dimensional results are compared with a three-dimensional finite element analysis based on reconstructed embryonic heart geometry. Through these steps, we found that the myocardium of zebrafish results in a stiffness on the order of 10 kPa immediately after the looping stage of development. This work enables the ability to determine how these properties change under normal and pathological heart development.
Collapse
Affiliation(s)
- Alex Gendernalik
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Room 304 Scott Building, 1374 Campus Delivery, Fort Collins, CO, 80523-1374, USA
| | - Neha Ahuja
- Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - Deborah Garrity
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA.,Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - David Bark
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA. .,Department of Mechanical Engineering, Colorado State University, Room 304 Scott Building, 1374 Campus Delivery, Fort Collins, CO, 80523-1374, USA. .,Department of Pediatrics, University of Colorado, Aurora, CO, USA. .,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
37
|
Kharkovskaya EE, Drugova OV, Osipov GV, Mukhina IV. Effect of Mechanical Stretching of the Right Atrium of Isolated Rat Heart on Dispersion of Repolarization before Fibrillation. Bull Exp Biol Med 2020; 169:438-440. [PMID: 32889569 DOI: 10.1007/s10517-020-04904-8] [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: 11/29/2019] [Indexed: 11/27/2022]
Abstract
The multi-electrode mapping method was used to analyze electrical activity of isolated rat heart under conditions of standard perfusion, pharmacological stimulation of fibrillation, and mechanical stretching of the right atrium both under normal conditions and before cardiac fibrillation. It was shown that stretching of the right atrium prevented the increase of repolarization dispersion and latency of the electrical signal in the myocardium that were observed before cardiac fibrillation.
Collapse
Affiliation(s)
- E E Kharkovskaya
- National Research N. I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| | - O V Drugova
- National Research N. I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - G V Osipov
- National Research N. I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - I V Mukhina
- National Research N. I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
38
|
Wang X, Senapati S, Akinbote A, Gnanasambandam B, Park PSH, Senyo SE. Microenvironment stiffness requires decellularized cardiac extracellular matrix to promote heart regeneration in the neonatal mouse heart. Acta Biomater 2020; 113:380-392. [PMID: 32590172 PMCID: PMC7428869 DOI: 10.1016/j.actbio.2020.06.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
The transient period of regeneration potential in the postnatal heart suggests molecular changes with maturation influence the cardiac response to damage. We have previously demonstrated that injury and exercise can stimulate cardiomyocyte proliferation in the adult heart suggesting a sensitivity to exogenous signals. Here, we consider whether exogenous fetal ECM and mechanically unloading interstitial matrix can drive regeneration after myocardial infarction (MI) surgery in low-regenerative hearts of day5 mice. Compared to controls, exogenous fetal ECM increases cardiac function and lowers fibrosis at 3 weeks post-injury and this effect can be augmented by softening heart tissue. In vitro experiments support a mechano-sensitivity to exogenous ECM signaling. We tested potential mechanisms and observed that fetal ECM increases nuclear YAP localization which could be enhanced by pharmacological stabilization of the cytoskeleton. Blocking YAP expression lowered fetal ECM effects though not completely. Lastly we observed mechanically unloading heart interstitial matrix increased agrin expression, an extracellular node in the YAP signaling pathway. Collectively, these data support a combined effect of exogenous factors and mechanical activity in altering agrin expression, cytoskeletal remodeling, and YAP signaling in driving cardiomyocyte cell cycle activity and regeneration in postnatal non-regenerative mice. STATEMENT OF SIGNIFICANCE: With the purpose of developing regenerative strategies, we investigate the influence of the local niche on the cardiac injury response. We conclude tissue stiffness, as anticipated in aging or disease, impairs regenerative therapeutics. Most novel, mechanical unloading facilitates enhanced cardiac regeneration only after cells are pushed into a permissive state by fetal biomolecules. Specifically, mechanical unloading appears to increase extracellular agrin expression that amplifies fetal-stimulation of nuclear YAP signaling which correlates with observed increases of cell cycle activity in cardiomyocytes. The results further suggest the cytoskeleton is critical to this interaction between mechanical unloading and independently actived YAP signaling. Using animal models, tissue explants, and cells, this work indicates that local mechanical stimuli can augment proliferating-permissive cardiomyocytes in the natural cardiac niche.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Subhadip Senapati
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University, United States
| | - Akinola Akinbote
- Department of Macromolecular Science & Engineering, Case Western Reserve University, United States
| | - Bhargavee Gnanasambandam
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Paul S-H Park
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University, United States
| | - Samuel E Senyo
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States.
| |
Collapse
|
39
|
VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci 2020; 21:ijms21155294. [PMID: 32722551 PMCID: PMC7432634 DOI: 10.3390/ijms21155294] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelial growth factor (VEGF), a homodimeric vasoactive glycoprotein, is the key mediator of angiogenesis. Angiogenesis, the formation of new blood vessels, is responsible for a wide variety of physio/pathological processes, including cardiovascular diseases (CVD). Cardiomyocytes (CM), the main cell type present in the heart, are the source and target of VEGF-A and express its receptors, VEGFR1 and VEGFR2, on their cell surface. The relationship between VEGF-A and the heart is double-sided. On the one hand, VEGF-A activates CM, inducing morphogenesis, contractility and wound healing. On the other hand, VEGF-A is produced by CM during inflammation, mechanical stress and cytokine stimulation. Moreover, high concentrations of VEGF-A have been found in patients affected by different CVD, and are often correlated with an unfavorable prognosis and disease severity. In this review, we summarized the current knowledge about the expression and effects of VEGF-A on CM and the role of VEGF-A in CVD, which are the most important cause of disability and premature death worldwide. Based on clinical studies on angiogenesis therapy conducted to date, it is possible to think that the control of angiogenesis and VEGF-A can lead to better quality and span of life of patients with heart disease.
Collapse
|
40
|
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: 2.0] [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
|
41
|
Kim MS, Fleres B, Lovett J, Anfinson M, Samudrala SSK, Kelly LJ, Teigen LE, Cavanaugh M, Marquez M, Geurts AM, Lough JW, Mitchell ME, Fitts RH, Tomita-Mitchell A. Contractility of Induced Pluripotent Stem Cell-Cardiomyocytes With an MYH6 Head Domain Variant Associated With Hypoplastic Left Heart Syndrome. Front Cell Dev Biol 2020; 8:440. [PMID: 32656206 PMCID: PMC7324479 DOI: 10.3389/fcell.2020.00440] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is a clinically and anatomically severe form of congenital heart disease; however, its etiology remains largely unknown. We previously demonstrated that genetic variants in the MYH6 gene are significantly associated with HLHS. Additionally, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from an HLHS-affected family trio (affected parent, unaffected parent, affected proband) carrying an MYH6-R443P head domain variant demonstrated dysmorphic sarcomere structure and increased compensatory MYH7 expression. Analysis of iPSC-CMs derived from the HLHS trio revealed that only beta myosin heavy chain expression was observed in CMs carrying the MYH6-R443P variant after differentiation day 15 (D15). Functional assessments performed between D20-D23 revealed that MYH6-R443P variant CMs contracted more slowly (40 ± 2 vs. 47 ± 2 contractions/min, P < 0.05), shortened less (5.6 ± 0.5 vs. 8.1 ± 0.7% of cell length, P < 0.05), and exhibited slower shortening rates (19.9 ± 1.7 vs. 28.1 ± 2.5 μm/s, P < 0.05) and relaxation rates (11.0 ± 0.9 vs. 19.7 ± 2.0 μm/s, P < 0.05). Treatment with isoproterenol had no effect on iPSC-CM mechanics. Using CRISPR/Cas9 gene editing technology, introduction of the R443P variant into the unaffected parent's iPSCs recapitulated the phenotype of the proband's iPSC-CMs, and conversely, correction of the R443P variant in the proband's iPSCs rescued the cardiomyogenic differentiation, sarcomere organization, slower contraction (P < 0.05) and decreased velocity phenotypes (P < 0.0001). This is the first report to identify that cardiac tissues from HLHS patients with MYH6 variants can exhibit sarcomere disorganization in atrial but not ventricular tissues. This new discovery was not unexpected, since MYH6 is expressed predominantly in the postnatal atria in humans. These findings demonstrate the feasibility of employing patient-derived iPSC-CMs, in combination with patient cardiac tissues, to gain mechanistic insight into how genetic variants can lead to HLHS. Results from this study suggest that decreased contractility of CMs due to sarcomere disorganization in the atria may effect hemodynamic changes preventing development of a normal left ventricle.
Collapse
Affiliation(s)
- Min-Su Kim
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Medical College of Wisconsin, Herma Heart Institute, Milwaukee, WI, United States
| | - Brandon Fleres
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Jerrell Lovett
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Melissa Anfinson
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sai Suma K Samudrala
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lauren J Kelly
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Laura E Teigen
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Matthew Cavanaugh
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Maribel Marquez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John W Lough
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael E Mitchell
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Medical College of Wisconsin, Herma Heart Institute, Milwaukee, WI, United States
| | - Robert H Fitts
- Department of Biological Sciences, Marquette University, Milwaukee, WI, United States
| | - Aoy Tomita-Mitchell
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Medical College of Wisconsin, Herma Heart Institute, Milwaukee, WI, United States.,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
42
|
Bousalis D, Lacko CS, Hlavac N, Alkassis F, Wachs RA, Mobini S, Schmidt CE, Kasahara H. Extracellular Matrix Disparities in an Nkx2-5 Mutant Mouse Model of Congenital Heart Disease. Front Cardiovasc Med 2020; 7:93. [PMID: 32548129 PMCID: PMC7272573 DOI: 10.3389/fcvm.2020.00093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/29/2020] [Indexed: 12/19/2022] Open
Abstract
Congenital heart disease (CHD) affects almost one percent of all live births. Despite diagnostic and surgical reparative advances, the causes and mechanisms of CHD are still primarily unknown. The extracellular matrix plays a large role in cell communication, function, and differentiation, and therefore likely plays a role in disease development and pathophysiology. Cell adhesion and gap junction proteins, such as integrins and connexins, are also essential to cellular communication and behavior, and could interact directly (integrins) or indirectly (connexins) with the extracellular matrix. In this work, we explore disparities in the expression and spatial patterning of extracellular matrix, adhesion, and gap junction proteins between wild type and Nkx2-5 +/R52G mutant mice. Decellularization and proteomic analysis, Western blotting, histology, immunostaining, and mechanical assessment of embryonic and neonatal wild type and Nkx2-5 mutant mouse hearts were performed. An increased abundance of collagen IV, fibronectin, and integrin β-1 was found in Nkx2-5 mutant neonatal mouse hearts, as well as increased expression of connexin 43 in embryonic mutant hearts. Furthermore, a ventricular noncompaction phenotype was observed in both embryonic and neonatal mutant hearts, as well as spatial disorganization of ECM proteins collagen IV and laminin in mutant hearts. Characterizing such properties in a mutant mouse model provides valuable information that can be applied to better understanding the mechanisms of congenital heart disease.
Collapse
Affiliation(s)
- Deanna Bousalis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Christopher S Lacko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Nora Hlavac
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Fariz Alkassis
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Rebecca A Wachs
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM+CSIC), Madrid, Spain.,Centro de Biología Molecular Severo Ochoa (CBMSO, UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
43
|
Sun S, Shi H, Moore S, Wang C, Ash-Shakoor A, Mather PT, Henderson JH, Ma Z. Progressive Myofibril Reorganization of Human Cardiomyocytes on a Dynamic Nanotopographic Substrate. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21450-21462. [PMID: 32326701 DOI: 10.1021/acsami.0c03464] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cardiomyocyte (CM) alignment with striated myofibril organization is developed during early cardiac organogenesis. Previous work has successfully achieved in vitro CM alignment using a variety of biomaterial scaffolds and substrates with static topographic features. However, the cellular processes that occur during the response of CMs to dynamic surface topographic changes, which may provide a model of in vivo developmental progress of CM alignment within embryonic myocardium, remains poorly understood. To gain insights into these cellular processes involved in the response of CMs to dynamic topographic changes, we developed a dynamic topographic substrate that employs a shape memory polymer coated with polyelectrolyte multilayers to produce a flat-to-wrinkle surface transition when triggered by a change in incubation temperature. Using this system, we investigated cellular morphological alignment and intracellular myofibril reorganization in response to the dynamic wrinkle formation. Hence, we identified the progressive cellular processes of human-induced pluripotent stem cell-CMs in a time-dependent manner, which could provide a foundation for a mechanistic model of cardiac myofibril reorganization in response to extracellular microenvironment changes.
Collapse
Affiliation(s)
- Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarah Moore
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Patrick T Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, Pennsylvania 17837, United States
| | - James H Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
44
|
Wang C, Koo S, Park M, Vangelatos Z, Hoang P, Conklin B, Grigoropoulos CP, Healy KE, Ma Z. Maladaptive Contractility of 3D Human Cardiac Microtissues to Mechanical Nonuniformity. Adv Healthc Mater 2020; 9:e1901373. [PMID: 32090507 PMCID: PMC7274862 DOI: 10.1002/adhm.201901373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/01/2020] [Indexed: 01/29/2023]
Abstract
Cardiac tissues are able to adjust their contractile behavior to adapt to the local mechanical environment. Nonuniformity of the native tissue mechanical properties contributes to the development of heart dysfunctions, yet the current in vitro cardiac tissue models often fail to recapitulate the mechanical nonuniformity. To address this issue, a 3D cardiac microtissue model is developed with engineered mechanical nonuniformity, enabled by 3D-printed hybrid matrices composed of fibers with different diameters. When escalating the complexity of tissue mechanical environments, cardiac microtissues start to develop maladaptive hypercontractile phenotypes, demonstrated in both contractile motion analysis and force-power analysis. This novel hybrid system could potentially facilitate the establishment of "pathologically-inspired" cardiac microtissue models for deeper understanding of heart pathology due to nonuniformity of the tissue mechanical environment.
Collapse
Affiliation(s)
- Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| | - Sangmo Koo
- Department of Mechanical Engineering, University of California, Berkeley
| | - Minok Park
- Department of Mechanical Engineering, University of California, Berkeley
| | | | - Plansky Hoang
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| | - Bruce Conklin
- Gladstone Institute of Cardiovascular Diseases, University of California, San Francisco
| | | | - Kevin E. Healy
- Department of Bioengineering, University of California, Berkeley
- Department of Material Science & Engineering, University of California, Berkeley
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| |
Collapse
|
45
|
Khokhlova A, Konovalov P, Iribe G, Solovyova O, Katsnelson L. The Effects of Mechanical Preload on Transmural Differences in Mechano-Calcium-Electric Feedback in Single Cardiomyocytes: Experiments and Mathematical Models. Front Physiol 2020; 11:171. [PMID: 32256377 PMCID: PMC7091561 DOI: 10.3389/fphys.2020.00171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/13/2020] [Indexed: 11/13/2022] Open
Abstract
Transmural differences in ventricular myocardium are maintained by electromechanical coupling and mechano-calcium/mechano-electric feedback. In the present study, we experimentally investigated the influence of preload on the force characteristics of subendocardial (Endo) and subepicardial (Epi) single ventricular cardiomyocytes stretched by up to 20% from slack sarcomere length (SL) and analyzed the results with the help of mathematical modeling. Mathematical models of Endo and Epi cells, which accounted for regional heterogeneity in ionic currents, Ca2+ handling, and myofilament contractile mechanisms, showed that a greater slope of the active tension–length relationship observed experimentally in Endo cardiomyocytes could be explained by greater length-dependent Ca2+ activation in Endo cells compared with Epi ones. The models also predicted that greater length dependence of Ca2+ activation in Endo cells compared to Epi ones underlies, via mechano-calcium-electric feedback, the reduction in the transmural gradient in action potential duration (APD) at a higher preload. However, the models were unable to reproduce the experimental data on a decrease of the transmural gradient in the time to peak contraction between Endo and Epi cells at longer end-diastolic SL. We hypothesize that preload-dependent changes in viscosity should be involved alongside the Frank–Starling effects to regulate the transmural gradient in length-dependent changes in the time course of contraction of Endo and Epi cardiomyocytes. Our experimental data and their analysis based on mathematical modeling give reason to believe that mechano-calcium-electric feedback plays a critical role in the modulation of electrophysiological and contractile properties of myocytes across the ventricular wall.
Collapse
Affiliation(s)
- Anastasia Khokhlova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia.,Institute of Natural Sciences and Mathematics, Ural Federal University, Yekaterinburg, Russia
| | - Pavel Konovalov
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia
| | - Gentaro Iribe
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan.,Department of Cardiovascular Physiology, Okayama University, Okayama, Japan
| | - Olga Solovyova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia.,Institute of Natural Sciences and Mathematics, Ural Federal University, Yekaterinburg, Russia
| | - Leonid Katsnelson
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, Russia.,Institute of Natural Sciences and Mathematics, Ural Federal University, Yekaterinburg, Russia
| |
Collapse
|
46
|
Sui Y, Zhang W, Tang T, Gao L, Cao T, Zhu H, You Q, Yu B, Yang T. Insulin-like growth factor-II overexpression accelerates parthenogenetic stem cell differentiation into cardiomyocytes and improves cardiac function after acute myocardial infarction in mice. Stem Cell Res Ther 2020; 11:86. [PMID: 32102690 PMCID: PMC7045450 DOI: 10.1186/s13287-020-1575-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/01/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Parthenogenetic stem cells (PSCs) are a promising source of regenerated cardiomyocytes; however, their application may be limited without a paternal genome. Insulin-like growth factor-II (IGF-II), a paternally expressed growth hormone, is critical in embryonic differentiation. This study investigated whether forced expression of IGF-II in PSCs can accelerate their differentiation. Methods Overexpression and re-knockdown of IGF-II in PSCs were performed to investigate the role of IGF-II in PSC differentiation. The derivatives of PSCs with different IGF-II manipulations were transplanted into infarcted murine hearts to investigate the role of IGF-II in cardiomyocyte differentiation in vivo. Results Data showed that the expression of cardiac troponin T and troponin I in IGF-II-PSC outgrowths preceded that of parental PSC outgrowths, suggesting that IGF-II can accelerate PSC differentiation into cardiac lineage. Overexpression of IGF-II accelerated PSC differentiation towards cardiomyocytes while inhibiting PSC proliferation via the IGF-II/IGF1R signaling. Similar to that observed in cardiac marker expression, on differentiation day 24, IGF-II-PSCs showed PCNA and cyclin D2 expression comparable to juvenile mouse cardiomyocytes, showing that IGF-II-PSCs at this stage possess differential and proliferative properties similar to those of juvenile cardiomyocytes. Moreover, the expression pattern of cardiac markers in IGF-II-overexpressing PSC derivatives resembled that of juvenile mouse cardiomyocytes. After transplantation into the infarcted mouse hearts, IGF-II-PSC-derived cardiomyocytes displayed significant characteristics of mature cardiomyocytes, and IGF-II-depletion by shRNA significantly reversed these effects, suggesting the critical role of IGF-II in promoting cardiomyocyte maturation in vivo. Furthermore, IGF-II-overexpressing PSC derivatives reduced collagen deposition and mitochondrial damage in the infarcted areas and improved cardiac function. The re-knockdown of IGF-II could counteract these favorable effects of IGF-II. Conclusions These findings suggest that the ectopic expression of IGF-II accelerates PSC differentiation into the cardiac lineage and promotes cardiomyocyte maturation. The underlying process includes the IGF-II/IGF1R signaling, which is involved in the suppressive effect of IGF-II on PSC proliferation. Moreover, transplanting IGF-II-overexpressing PSC derivatives into the infarcted heart could reduce collagen deposition and improve mitochondria biogenesis and measurements of cardiac function, highlighting the importance of IGF-II in the application of PSCs in cardiac regeneration.
Collapse
Affiliation(s)
- Yi Sui
- Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Zhang
- Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Tao Tang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Lili Gao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Ting Cao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hongbo Zhu
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Bo Yu
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
47
|
Verjans R, van Bilsen M, Schroen B. Reviewing the Limitations of Adult Mammalian Cardiac Regeneration: Noncoding RNAs as Regulators of Cardiomyogenesis. Biomolecules 2020; 10:biom10020262. [PMID: 32050588 PMCID: PMC7072544 DOI: 10.3390/biom10020262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 12/18/2022] Open
Abstract
The adult mammalian heart is incapable of regeneration following cardiac injury, leading to a decline in function and eventually heart failure. One of the most evident barriers limiting cardiac regeneration is the inability of cardiomyocytes to divide. It has recently become clear that the mammalian heart undergoes limited cardiomyocyte self-renewal throughout life and is even capable of modest regeneration early after birth. These exciting findings have awakened the goal to promote cardiomyogenesis of the human heart to repair cardiac injury or treat heart failure. We are still far from understanding why adult mammalian cardiomyocytes possess only a limited capacity to proliferate. Identifying the key regulators may help to progress towards such revolutionary therapy. Specific noncoding RNAs control cardiomyocyte division, including well explored microRNAs and more recently emerged long noncoding RNAs. Elucidating their function and molecular mechanisms during cardiomyogenesis is a prerequisite to advance towards therapeutic options for cardiac regeneration. In this review, we present an overview of the molecular basis of cardiac regeneration and describe current evidence implicating microRNAs and long noncoding RNAs in this process. Current limitations and future opportunities regarding how these regulatory mechanisms can be harnessed to study myocardial regeneration will be addressed.
Collapse
Affiliation(s)
- Robin Verjans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Marc van Bilsen
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Blanche Schroen
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
- Correspondence: ; Tel.: +31-433882949
| |
Collapse
|
48
|
Karbassi E, Fenix A, Marchiano S, Muraoka N, Nakamura K, Yang X, Murry CE. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol 2020; 17:341-359. [PMID: 32015528 DOI: 10.1038/s41569-019-0331-x] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Our knowledge of pluripotent stem cell (PSC) biology has advanced to the point where we now can generate most cells of the human body in the laboratory. PSC-derived cardiomyocytes can be generated routinely with high yield and purity for disease research and drug development, and these cells are now gradually entering the clinical research phase for the testing of heart regeneration therapies. However, a major hurdle for their applications is the immature state of these cardiomyocytes. In this Review, we describe the structural and functional properties of cardiomyocytes and present the current approaches to mature PSC-derived cardiomyocytes. To date, the greatest success in maturation of PSC-derived cardiomyocytes has been with transplantation into the heart in animal models and the engineering of 3D heart tissues with electromechanical conditioning. In conventional 2D cell culture, biophysical stimuli such as mechanical loading, electrical stimulation and nanotopology cues all induce substantial maturation, particularly of the contractile cytoskeleton. Metabolism has emerged as a potent means to control maturation with unexpected effects on electrical and mechanical function. Different interventions induce distinct facets of maturation, suggesting that activating multiple signalling networks might lead to increased maturation. Despite considerable progress, we are still far from being able to generate PSC-derived cardiomyocytes with adult-like phenotypes in vitro. Future progress will come from identifying the developmental drivers of maturation and leveraging them to create more mature cardiomyocytes for research and regenerative medicine.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Aidan Fenix
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Silvia Marchiano
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Naoto Muraoka
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA. .,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA. .,Department of Pathology, University of Washington, Seattle, WA, USA. .,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
49
|
Alassaf A, Tansik G, Mayo V, Wubker L, Carbonero D, Agarwal A. Engineering anisotropic cardiac monolayers on microelectrode arrays for non-invasive analyses of electrophysiological properties. Analyst 2020; 145:139-149. [DOI: 10.1039/c9an01339c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Engineering cardiac tissues with physiological architectural and mechanical properties on microelectrode arrays enables long term culture and non-invasive collection of electrophysiological readouts.
Collapse
Affiliation(s)
- Ahmad Alassaf
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Department of Medical Equipment Technology
| | - Gulistan Tansik
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Vera Mayo
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Laura Wubker
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Daniel Carbonero
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Dr. John T Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami
| |
Collapse
|
50
|
Gomes A, Oudot C, Macià A, Foito A, Carregosa D, Stewart D, Van de Wiele T, Berry D, Motilva MJ, Brenner C, Dos Santos CN. Berry-Enriched Diet in Salt-Sensitive Hypertensive Rats: Metabolic Fate of (Poly)Phenols and the Role of Gut Microbiota. Nutrients 2019; 11:E2634. [PMID: 31684148 PMCID: PMC6893819 DOI: 10.3390/nu11112634] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023] Open
Abstract
Diets rich in (poly)phenols are associated with a reduced reduction in the incidence of cardiovascular disorders. While the absorption and metabolism of (poly)phenols has been described, it is not clear how their metabolic fate is affected under pathological conditions. This study evaluated the metabolic fate of berry (poly)phenols in an in vivo model of hypertension as well as the associated microbiota response. Dahl salt-sensitive rats were fed either a low-salt diet (0.26% NaCl) or a high-salt diet (8% NaCl), with or without a berry mixture (blueberries, blackberries, raspberries, Portuguese crowberry and strawberry tree fruit) for 9 weeks. The salt-enriched diet promoted an increase in the urinary excretion of berry (poly)phenol metabolites, while the abundance of these metabolites decreased in faeces, as revealed by UPLC-MS/MS. Moreover, salt and berries modulated gut microbiota composition as demonstrated by 16S rRNA analysis. Some changes in the microbiota composition were associated with the high-salt diet and revealed an expansion of the families Proteobacteria and Erysipelotrichaceae. However, this effect was mitigated by the dietary supplementation with berries. Alterations in the metabolic fate of (poly)phenols occur in parallel with the modulation of gut microbiota in hypertensive rats. Thus, beneficial effects of (poly)phenols could be related with these interlinked modifications, between metabolites and microbiota environments.
Collapse
Affiliation(s)
- Andreia Gomes
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| | - Carole Oudot
- INSERM UMR-S 1180- University Paris-Sud, University Paris Saclay, 5 rue Jean-Baptiste Clément, 92296 Châtenay Malabry, France.
| | - Alba Macià
- Food Technology Department, Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, University of Lleida, 25198-Lleida, Spain.
| | - Alexandre Foito
- Environmental and Biochemical Sciences, James Hutton Institute, Invergowrie Dundee DD2 5DA Scotland, UK.
| | - Diogo Carregosa
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
| | - Derek Stewart
- Environmental and Biochemical Sciences, James Hutton Institute, Invergowrie Dundee DD2 5DA Scotland, UK.
- Institute of Mechanical Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS Scotland, UK.
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Research Network Chemistry Meets Microbiology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| | - Maria-José Motilva
- Instituto de Ciencias de la Vid y del Vino-ICVV, CSIC-Universidad de La Rioja-Gobierno de La Rioja, Finca "La Grajera", Carretera de Burgos km 6, 26007 Logroño, Spain.
| | - Catherine Brenner
- INSERM UMR-S 1180- University Paris-Sud, University Paris Saclay, 5 rue Jean-Baptiste Clément, 92296 Châtenay Malabry, France.
| | - Cláudia Nunes Dos Santos
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
| |
Collapse
|