1
|
Shang L, Wang S, Mao Y. Recent advances in plant-derived polysaccharide scaffolds in tissue engineering: A review. Int J Biol Macromol 2024; 277:133830. [PMID: 39002914 DOI: 10.1016/j.ijbiomac.2024.133830] [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: 03/03/2024] [Revised: 06/13/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
As a natural three-dimensional biopolymer, decellularized plant-derived scaffolds usually comprise various polysaccharides, mostly cellulose, pectin, and hemicellulose. They are characterized by natural biocompatibility and porous structures. The emergence of decellularized purified polysaccharide scaffolds provides an attractive method to overcome the challenges associated with nutrient delivery and biocompatibility, as they serve as optimal non-immune environments for stem cell adhesion and proliferation. To date, limited corresponding literature is available to systemically summarize the development and potential of these scaffolds in tissue engineering. Therefore, the current review summarized the biomimetic properties of plant-derived polysaccharide scaffolds and the latest progress in tissue engineering applications. This review first discusses the advantages of decellularized plant-derived polysaccharide scaffolds by briefly introducing their features and current limitations in clinical applications. Subsequently, the latest progress in emerging applications of regenerative biomaterials is reviewed, followed by a discussion of the studies on the interactions of biomaterials with cells and tissues. Finally, challenges in obtaining reliable scaffolds and possible future directions are discussed.
Collapse
Affiliation(s)
- Lijun Shang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Shan Wang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
2
|
Chen TA, Zhao BB, Balbin RA, Sharma S, Ha D, Kamp TJ, Zhou Y, Zhao F. Engineering a robust and anisotropic cardiac-specific extracellular matrix scaffold for cardiac patch tissue engineering. Matrix Biol Plus 2024; 23:100151. [PMID: 38882397 PMCID: PMC11176808 DOI: 10.1016/j.mbplus.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/22/2024] [Accepted: 05/18/2024] [Indexed: 06/18/2024] Open
Abstract
Extracellular matrix (ECM) fabricated using human induced pluripotent stem cells (hiPSCs)-derived cardiac fibroblasts (hiPSC-CFs) could serve as a completely biological scaffold for an engineered cardiac patch, leveraging the unlimited source and outstanding reproducibility of hiPSC-CFs. Additionally, hiPSC-CF-derived ECM (hiPSC-CF-ECM) holds the potential to enhance maturation of exogenous cardiomyocytes, such as hiPSC-derived cardiomyocytes (hiPSC-CMs), by providing a microenvironment rich in cardiac-specific biochemical and signaling cues. However, achieving sufficient robustness of hiPSC-CF-ECM is challenging. This study aims to achieve appropriate ECM deposition, scaffold thickness, and mechanical strength of an aligned hiPSC-CF-ECM by optimizing the culture period, ranging from 2 to 10 weeks, of hiPSC-CFs grown on micro-grated substrates, which can direct the alignment of both hiPSC-CFs and their secreted ECM. The hiPSC-CFs demonstrated a production rate of 13.5 µg ECM per day per 20,000 cells seeded. An anisotropic nanofibrous hiPSC-CF-ECM scaffold with a thickness of 20.0 ± 2.1 µm was achieved after 6 weeks of culture, followed by decellularization. Compositional analysis through liquid chromatography-mass spectrometry (LC-MS) revealed the presence of cardiac-specific fibrillar collagens, non-fibrillar collagens, and matricellular proteins. Uniaxial tensile stretching of the hiPSC-CF-ECM scaffold indicated robust tensile resilience. Finally, hiPSCs-CMs cultured on the hiPSC-CF-ECM exhibited alignment following the guidance of ECM nanofibers and demonstrated mature organization of key structural proteins. The culture duration of the anisotropic hiPSC-CF-ECM was successfully refined to achieve a robust scaffold containing structural proteins that resembles cardiac microenvironment. This completely biological, anisotropic, and cardiac-specific ECM holds great potential for cardiac patch engineering.
Collapse
Affiliation(s)
- Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Brandon B. Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Richard A. Balbin
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Sameeksha Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Donggi Ha
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Timothy J. Kamp
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
3
|
Wang D, Zhang H, Chen Y, He J, Zhao L, Huang Y, Zhao F, Jiang Y, Fu S, Hong Z. Improving therapeutic effects of exosomes encapsulated gelatin methacryloyl/hyaluronic acid blended and oxygen releasing injectable hydrogel by cardiomyocytes induction and vascularization in rat myocardial infarction model. Int J Biol Macromol 2024; 271:132412. [PMID: 38754674 DOI: 10.1016/j.ijbiomac.2024.132412] [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: 11/07/2023] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/18/2024]
Abstract
Acute myocardial infarction (AMI) causes acute cardiac cell death when oxygen supply is disrupted. Improving oxygen flow to the damaged area could potentially achieve the to prevent cell death and provide cardiac regeneration. Here, we describe the production of oxygen-producing injectable bio-macromolecular hydrogels from natural polymeric components including gelatin methacryloyl (GelMA), hyaluronic acid (HA) loaded with catalase (CAT). Under hypoxic conditions, the O2-generating hydrogels (O2 (+) hydrogel) encapsulated with Mesenchymal stem cells (MSCs)-derived-exosomes (Exo- O2 (+) hydrogel) released substantial amounts of oxygen for >5 days. We demonstrated that after 7 days of in vitro cell culture, exhibits identical production of paracrine factors compared to those of culture of rat cardiac fibroblasts (RCFs), rat neonatal cardiomyocytes (RNCs) and Human Umbilical Vein Endothelial Cells (HUVECs), demonstrating its ability to replicate the natural architecture and function of capillaries. Four weeks after treatment with Exo-O2 (+) hydrogel, cardiomyocytes in the peri-infarct area of an in vivo rat model of AMI displayed substantial mitotic activity. In contrast with infarcted hearts treated with O2 (-) hydrogel, Exo- O2 (+) hydrogel infarcted hearts showed a considerable increase in myocardial capillary density. The outstanding therapeutic advantages and quick, easy fabrication of Exo- O2 (+) hydrogel has provided promise favourably for potential cardiac treatment applications.
Collapse
Affiliation(s)
- Dan Wang
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Hong Zhang
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yu Chen
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Jiangchun He
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Li Zhao
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yixiong Huang
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Fengjiao Zhao
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yuting Jiang
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Shihu Fu
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China.
| | - Zhibo Hong
- Department of Cardiology, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China.
| |
Collapse
|
4
|
Yao ZF, Kuang Y, Wu HT, Lundqvist E, Fu X, Celt N, Pei J, Yee A, Ardoña HAM. Selective Induction of Molecular Assembly to Tissue-Level Anisotropy on Peptide-Based Optoelectronic Cardiac Biointerfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312231. [PMID: 38335948 PMCID: PMC11126358 DOI: 10.1002/adma.202312231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/19/2024] [Indexed: 02/12/2024]
Abstract
The conduction efficiency of ions in excitable tissues and of charged species in organic conjugated materials both benefit from having ordered domains and anisotropic pathways. In this study, a photocurrent-generating cardiac biointerface is presented, particularly for investigating the sensitivity of cardiomyocytes to geometrically comply to biomacromolecular cues differentially assembled on a conductive nanogrooved substrate. Through a polymeric surface-templated approach, photoconductive substrates with symmetric peptide-quaterthiophene (4T)-peptide units assembled as 1D nanostructures on nanoimprinted polyalkylthiophene (P3HT) surface are developed. The 4T-based peptides studied here can form 1D nanostructures on prepatterned polyalkylthiophene substrates, as directed by hydrogen bonding, aromatic interactions between 4T and P3HT, and physical confinement on the nanogrooves. It is observed that smaller 4T-peptide units that can achieve a higher degree of assembly order within the polymeric templates serve as a more efficient driver of cardiac cytoskeletal anisotropy than merely presenting aligned -RGD bioadhesive epitopes on a nanotopographic surface. These results unravel some insights on how cardiomyocytes perceive submicrometer dimensionality, local molecular order, and characteristics of surface cues in their immediate environment. Overall, the work offers a cardiac patterning platform that presents the possibility of a gene modification-free cardiac photostimulation approach while controlling the conduction directionality of the biotic and abiotic components.
Collapse
Affiliation(s)
- Ze-Fan Yao
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
| | - Yuyao Kuang
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Hao-Tian Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Emil Lundqvist
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Xin Fu
- Department of Materials Science and Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Natalie Celt
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Jian Pei
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Albert Yee
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Herdeline Ann M. Ardoña
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
5
|
Park S, Laskow TC, Chen J, Guha P, Dawn B, Kim D. Microphysiological systems for human aging research. Aging Cell 2024; 23:e14070. [PMID: 38180277 PMCID: PMC10928588 DOI: 10.1111/acel.14070] [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: 06/30/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in microphysiological systems (MPS), also known as organs-on-a-chip (OoC), enable the recapitulation of more complex organ and tissue functions on a smaller scale in vitro. MPS therefore provide the potential to better understand human diseases and physiology. To date, numerous MPS platforms have been developed for various tissues and organs, including the heart, liver, kidney, blood vessels, muscle, and adipose tissue. However, only a few studies have explored using MPS platforms to unravel the effects of aging on human physiology and the pathogenesis of age-related diseases. Age is one of the risk factors for many diseases, and enormous interest has been devoted to aging research. As such, a human MPS aging model could provide a more predictive tool to understand the molecular and cellular mechanisms underlying human aging and age-related diseases. These models can also be used to evaluate preclinical drugs for age-related diseases and translate them into clinical settings. Here, we provide a review on the application of MPS in aging research. First, we offer an overview of the molecular, cellular, and physiological changes with age in several tissues or organs. Next, we discuss previous aging models and the current state of MPS for studying human aging and age-related conditions. Lastly, we address the limitations of current MPS and present future directions on the potential of MPS platforms for human aging research.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical EngineeringUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Thomas C. Laskow
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jingchun Chen
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Prasun Guha
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
- School of Life SciencesUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Deok‐Ho Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
6
|
Verdonk SJE, Storoni S, Micha D, van den Aardweg JG, Versacci P, Celli L, de Vries R, Zhytnik L, Kamp O, Bugiani M, Eekhoff EMW. Is Osteogenesis Imperfecta Associated with Cardiovascular Abnormalities? A Systematic Review of the Literature. Calcif Tissue Int 2024; 114:210-221. [PMID: 38243143 PMCID: PMC10902066 DOI: 10.1007/s00223-023-01171-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/27/2023] [Indexed: 01/21/2024]
Abstract
Osteogenesis imperfecta (OI) is a rare genetic disorder caused by abnormal collagen type I production. While OI is primarily characterized by bone fragility and deformities, patients also have extraskeletal manifestations, including an increased risk of cardiovascular disease. This review provides a comprehensive overview of the literature on cardiovascular diseases in OI patients in order to raise awareness of this understudied clinical aspect of OI and support clinical guidelines. In accordance with the PRISMA guidelines, a systematic literature search in PubMed, Embase, Web of Science and Scopus was conducted that included articles from the inception of these databases to April 2023. Valvular disease, heart failure, atrial fibrillation, and hypertension appear to be more prevalent in OI than in control individuals. Moreover, a larger aortic root was observed in OI compared to controls. Various cardiovascular diseases appear to be more prevalent in OI than in controls. These cardiovascular abnormalities are observed in all types of OI and at all ages, including young children. As there are insufficient longitudinal studies, it is unknown whether these abnormalities are progressive in nature in OI patients. Based on these findings, we would recommend referring individuals with OI to a cardiologist with a low-threshold.
Collapse
Affiliation(s)
- Sara J E Verdonk
- Department of Endocrinology and Metabolism, Amsterdam UMC Location Vrije Universiteit, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Rare Bone Disease Center, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Silvia Storoni
- Department of Endocrinology and Metabolism, Amsterdam UMC Location Vrije Universiteit, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Rare Bone Disease Center, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Dimitra Micha
- Rare Bone Disease Center, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Joost G van den Aardweg
- Department of Respiratory Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Paolo Versacci
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Luca Celli
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lidiia Zhytnik
- Rare Bone Disease Center, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Department of Traumatology and Orthopeadics, University of Tartu, Tartu, Estonia
| | - Otto Kamp
- Department of Cardiology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
| | - Elisabeth M W Eekhoff
- Department of Endocrinology and Metabolism, Amsterdam UMC Location Vrije Universiteit, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Rare Bone Disease Center, Amsterdam, The Netherlands.
- Amsterdam Movement Sciences, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Perreault LR, Daley MC, Watson MC, Rastogi S, Jaiganesh A, Porter EC, Duffy BM, Black LD. Characterization of cardiac fibroblast-extracellular matrix crosstalk across developmental ages provides insight into age-related changes in cardiac repair. Front Cell Dev Biol 2024; 12:1279932. [PMID: 38434619 PMCID: PMC10904575 DOI: 10.3389/fcell.2024.1279932] [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: 08/18/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Heart failure afflicts an estimated 6.5 million people in the United States, driven largely by incidents of coronary heart disease (CHD). CHD leads to heart failure due to the inability of adult myocardial tissue to regenerate after myocardial infarction (MI). Instead, immune cells and resident cardiac fibroblasts (CFs), the cells responsible for the maintenance of the cardiac extracellular matrix (cECM), drive an inflammatory wound healing response, which leads to fibrotic scar tissue. However, fibrosis is reduced in fetal and early (<1-week-old) neonatal mammals, which exhibit a transient capability for regenerative tissue remodeling. Recent work by our laboratory and others suggests this is in part due to compositional differences in the cECM and functional differences in CFs with respect to developmental age. Specifically, fetal cECM and CFs appear to mitigate functional loss in MI models and engineered cardiac tissues, compared to adult CFs and cECM. We conducted 2D studies of CFs on solubilized fetal and adult cECM to investigate whether these age-specific functional differences are synergistic with respect to their impact on CF phenotype and, therefore, cardiac wound healing. We found that the CF migration rate and stiffness vary with respect to cell and cECM developmental age and that CF transition to a fibrotic phenotype can be partially attenuated in the fetal cECM. However, this effect was not observed when cells were treated with cytokine TGF-β1, suggesting that inflammatory signaling factors are the dominant driver of the fibroblast phenotype. This information may be valuable for targeted therapies aimed at modifying the CF wound healing response and is broadly applicable to age-related studies of cardiac remodeling.
Collapse
Affiliation(s)
- Luke R. Perreault
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Mark C. Daley
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Matthew C. Watson
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Sagar Rastogi
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Ajith Jaiganesh
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Elizabeth C. Porter
- Cellular, Molecular and Developmental Biology Program, Graduate School for Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Breanna M. Duffy
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Lauren D. Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Cellular, Molecular and Developmental Biology Program, Graduate School for Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
8
|
Hamsho K, Broadwin M, Stone CR, Sellke FW, Abid MR. The Current State of Extracellular Matrix Therapy for Ischemic Heart Disease. Med Sci (Basel) 2024; 12:8. [PMID: 38390858 PMCID: PMC10885030 DOI: 10.3390/medsci12010008] [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: 12/21/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
The extracellular matrix (ECM) is a three-dimensional, acellular network of diverse structural and nonstructural proteins embedded within a gel-like ground substance composed of glycosaminoglycans and proteoglycans. The ECM serves numerous roles that vary according to the tissue in which it is situated. In the myocardium, the ECM acts as a collagen-based scaffold that mediates the transmission of contractile signals, provides means for paracrine signaling, and maintains nutritional and immunologic homeostasis. Given this spectrum, it is unsurprising that both the composition and role of the ECM has been found to be modulated in the context of cardiac pathology. Myocardial infarction (MI) provides a familiar example of this; the ECM changes in a way that is characteristic of the progressive phases of post-infarction healing. In recent years, this involvement in infarct pathophysiology has prompted a search for therapeutic targets: if ECM components facilitate healing, then their manipulation may accelerate recovery, or even reverse pre-existing damage. This possibility has been the subject of numerous efforts involving the integration of ECM-based therapies, either derived directly from biologic sources or bioengineered sources, into models of myocardial disease. In this paper, we provide a thorough review of the published literature on the use of the ECM as a novel therapy for ischemic heart disease, with a focus on biologically derived models, of both the whole ECM and the components thereof.
Collapse
Affiliation(s)
- Khaled Hamsho
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.H.); (M.B.); (C.R.S.); (F.W.S.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.H.); (M.B.); (C.R.S.); (F.W.S.)
| | - Christopher R. Stone
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.H.); (M.B.); (C.R.S.); (F.W.S.)
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.H.); (M.B.); (C.R.S.); (F.W.S.)
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.H.); (M.B.); (C.R.S.); (F.W.S.)
| |
Collapse
|
9
|
Wang X, Kulik K, Wan TC, Lough JW, Auchampach JA. Evidence of Histone H2A.Z Deacetylation and Cardiomyocyte Dedifferentiation in Infarcted/Tip60-depleted Hearts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575312. [PMID: 38260622 PMCID: PMC10802610 DOI: 10.1101/2024.01.11.575312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Myocardial infarction (MI) in the human heart causes death of billions of cardiomyocytes (CMs), resulting in cardiac dysfunction that is incompatible with life or lifestyle. In order to re-muscularize injured myocardium, replacement CMs must be generated via renewed proliferation of surviving CMs. Approaches designed to induce proliferation of CMs after injury have been insufficient. Toward this end, we are targeting the Tip60 acetyltransferase, based on the rationale that its pleiotropic functions conspire to block the CM cell-cycle at several checkpoints. We previously reported that genetic depletion of Tip60 in a mouse model after MI reduces scarring, retains cardiac function, and activates the CM cell-cycle, although it is unclear whether this culminates in the generation of daughter CMs. For pre-existing CMs in the adult heart to resume proliferation, it is becoming widely accepted that they must first dedifferentiate, a process highlighted by loss of maturity, epithelial to mesenchymal transitioning (EMT), and reversion from fatty acid oxidation to glycolytic metabolism, accompanied by softening of the myocardial extracellular matrix. Findings in hematopoietic stem cells, and more recently in neural progenitor cells, have shown that Tip60 induces and maintains the differentiated state via site-specific acetylation of the histone variant H2A.Z. Here, we report that genetic depletion of Tip60 from naïve or infarcted hearts results in the near-complete absence of acetylated H2A.Z in CM nuclei, and that this is accordingly accompanied by altered gene expressions indicative of EMT induction, ECM softening, decreased fatty acid oxidation, and depressed expression of genes that regulate the TCA cycle. These findings, combined with our previous work, support the notion that because Tip60 has multiple targets that combinatorially maintain the differentiated state and inhibit proliferation, its transient therapeutic targeting to ameliorate the effects of cardiac injury should be considered.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin Milwaukee, WI 53226
| | - Katherine Kulik
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin Milwaukee, WI 53226
| | - Tina C. Wan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin Milwaukee, WI 53226
| | - John W. Lough
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin Milwaukee, WI 53226
| | - John A. Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin Milwaukee, WI 53226
| |
Collapse
|
10
|
Rubio LD, McFarland KA, O'Seaghdha M, Williams C. A high throughput microphysiological model of prosthetic valve endocarditis for investigating factors that influence bacterial adhesion under fluid shear stress. Biochem Biophys Res Commun 2023; 686:149155. [PMID: 37926046 DOI: 10.1016/j.bbrc.2023.149155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Prosthetic heart valves are associated with almost one quarter of cases of infective endocarditis, a rare but serious condition with a staggering 25 % mortality rate. Without the endothelium of native valves, the risk of infection is exacerbated for implanted devices exposed to blood. There are currently no physiologically relevant in vitro or animal models of prosthetic valve endocarditis (PVE). Of particular importance, Staphylococcus aureus, a common agent of PVE, has demonstrated enhanced binding to blood plasma proteins (e.g., fibrinogen) and exposed matrix under fluid shear stress (FSS). An in vitro platform that mimics the multiple physiological determinants for S. aureus adhesion to prosthetic valve materials would facilitate the discovery of new treatments to minimize PVE. To this end, we developed a first-of-its-kind microphysiological model of PVE to study the effects of several key variables (endothelial cell coverage, fibrinogen deposition, surface treatments, and FSS) on S. aureus adhesion to bioprosthetic material surfaces. Our model demonstrated that viable endothelial monolayers diminished the deposition of fibrinogen and that fibrinogen was required for the subsequent adhesion of S. aureus to the bioprosthetic surface model. Next, we examined factors that affected endothelial cell coverage, such as FSS and glutaraldehyde, a common chemical treatment for bioprosthetic materials. In particular, glutaraldehyde treatment obstructed endothelialization of otherwise biocompatible collagen-coated surfaces, further enabling fibrinogen and S. aureus deposition. In future work, this model could impact multiple research areas, such as screening candidate bioprosthetic valve materials and new surface treatments to prevent PVE and further understanding host-pathogen interactions.
Collapse
Affiliation(s)
- Logan D Rubio
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Kirsty A McFarland
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | | | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States.
| |
Collapse
|
11
|
Fernandes I, Funakoshi S, Hamidzada H, Epelman S, Keller G. Modeling cardiac fibroblast heterogeneity from human pluripotent stem cell-derived epicardial cells. Nat Commun 2023; 14:8183. [PMID: 38081833 PMCID: PMC10713677 DOI: 10.1038/s41467-023-43312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Cardiac fibroblasts play an essential role in the development of the heart and are implicated in disease progression in the context of fibrosis and regeneration. Here, we establish a simple organoid culture platform using human pluripotent stem cell-derived epicardial cells and ventricular cardiomyocytes to study the development, maturation, and heterogeneity of cardiac fibroblasts under normal conditions and following treatment with pathological stimuli. We demonstrate that this system models the early interactions between epicardial cells and cardiomyocytes to generate a population of fibroblasts that recapitulates many aspects of fibroblast behavior in vivo, including changes associated with maturation and in response to pathological stimuli associated with cardiac injury. Using single cell transcriptomics, we show that the hPSC-derived organoid fibroblast population displays a high degree of heterogeneity that approximates the heterogeneity of populations in both the normal and diseased human heart. Additionally, we identify a unique subpopulation of fibroblasts possessing reparative features previously characterized in the hearts of model organisms. Taken together, our system recapitulates many aspects of human cardiac fibroblast specification, development, and maturation, providing a platform to investigate the role of these cells in human cardiovascular development and disease.
Collapse
Affiliation(s)
- Ian Fernandes
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Shunsuke Funakoshi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G1L7, Canada.
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network Toronto, Toronto, ON, M5G1L7, Canada
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, M5G1L7, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5G1L7, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network Toronto, Toronto, ON, M5G1L7, Canada
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, M5G1L7, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5G1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G1L7, Canada
- Peter Munk Cardiac Centre, University Health Networ, Toronto, ON, M5G1L7, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G1L7, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada.
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G1L7, Canada.
| |
Collapse
|
12
|
Wang X, Shi H, Huang S, Zhang Y, He X, Long Q, Qian B, Zhong Y, Qi Z, Zhao Q, Ye X. Localized delivery of anti-inflammatory agents using extracellular matrix-nanostructured lipid carriers hydrogel promotes cardiac repair post-myocardial infarction. Biomaterials 2023; 302:122364. [PMID: 37883909 DOI: 10.1016/j.biomaterials.2023.122364] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023]
Abstract
A challenge in treating cardiac injury is the low heart-specificity of the drugs. Nanostructured lipid carriers (NLCs) are a relatively new format of lipid nanoparticles which have been used to deliver RNA and drugs. However, lipid nanoparticles exhibit higher affinity to the liver than the heart. To improve the delivery efficiency of NLCs into the heart, NLCs can be embedded into a scaffold and be locally released. In this study, a cardiac extracellular matrix (ECM) hydrogel-NLC composite was developed as a platform for cardiac repair. ECM-NLC composite gels at physiological conditions and releases payloads into the heart over weeks. ECM-NLC hydrogel carrying colchicine, an anti-inflammation agent, improved cardiac repair after myocardial infarction in mice. Transcriptome analysis indicated that Egfr downstream effectors participated in ECM-NLC-colchicine induced heart repair. In conclusion, ECM-NLC hydrogel is a potential platform for sustained and localized delivery of biomolecules into the heart, and loading appropriate medicines further increases the therapeutic efficacy of ECM-NLC hydrogel for cardiovascular diseases.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Hongpeng Shi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaojun He
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Ozcebe SG, Zorlutuna P. In need of age-appropriate cardiac models: Impact of cell age on extracellular matrix therapy outcomes. Aging Cell 2023; 22:e13966. [PMID: 37803909 PMCID: PMC10652343 DOI: 10.1111/acel.13966] [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: 03/13/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 10/08/2023] Open
Abstract
Aging is the main risk factor for cardiovascular disease (CVD). As the world's population ages rapidly and CVD rates rise, there is a growing need for physiologically relevant models of aging hearts to better understand cardiac aging. Translational research relies heavily on young animal models; however, these models correspond to early ages in human life, therefore cannot fully capture the pathophysiology of age-related CVD. Here, we first investigated the transcriptomic and proteomic changes that occur with human cardiac aging. We then chronologically aged human induced pluripotent stem cell-derived cardiomyocytes (iCMs) and showed that 14-month-old iCMs exhibited a similar aging profile to the human CMs and recapitulated age-related disease hallmarks. Using aged iCMs, we studied the effect of cell age on the young extracellular matrix (ECM) therapy, an emerging approach for myocardial infarction (MI) treatment and prevention. Young ECM decreased oxidative stress, improved survival, and post-MI beating in aged iCMs. In the absence of stress, young ECM improved beating and reversed aging-associated expressions in 3-month-old iCMs while causing the opposite effect on 14-month-old iCMs. The same young ECM treatment surprisingly increased SASP and impaired beating in advanced aged iCMs. Overall, we showed that young ECM therapy had a positive effect on post-MI recovery; however, cell age was determinant in the treatment outcomes without any stress conditions. Therefore, "one-size-fits-all" approaches to ECM treatments fail, and cardiac tissue engineered models with age-matched human iCMs are valuable in translational basic research for determining the appropriate treatment, particularly for the elderly.
Collapse
Affiliation(s)
- S. Gulberk Ozcebe
- Bioengineering Graduate ProgramUniversity of Notre DameNotre DameIndianaUSA
| | - Pinar Zorlutuna
- Bioengineering Graduate ProgramUniversity of Notre DameNotre DameIndianaUSA
- Department of Aerospace and Mechanical EngineeringUniversity of Notre DameNotre DameIndianaUSA
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
14
|
Kar A, Sable M, A A, Jena SK, Tripathy PR, Gaikwad M. An Immunohistochemical Study of Proliferation of Human Fetal Heart Cardiomyocyte With Phospho-Histone H3 Antibody. Cureus 2023; 15:e41159. [PMID: 37525760 PMCID: PMC10387164 DOI: 10.7759/cureus.41159] [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] [Accepted: 06/29/2023] [Indexed: 08/02/2023] Open
Abstract
The rapid proliferation of cardiomyocytes in mammals occurs during fetal life. But in postnatal life, this capacity of proliferation is reduced or lost as they exit the cell cycle. However, the cardiomyocytes don't show the same activity for different species. In human fetuses or in adult life, the capacity of the proliferation of cardiomyocytes and their response to an injury are not understood yet. In this study, we have done an immunohistochemical study using phospho-histone H3 (PHH3) to observe human fetal cardiomyocytes' proliferative activity. The heart specimens from the fetal autopsy of spontaneously aborted and stillborn human fetuses were subjected to immunohistochemical study using PHH3 antibody, and comparison between the PHH3 index (number of PHH3 positive cells per 1000 number of cardiomyocytes/high power field [HPF]) of myocardial regions was done using appropriate statistical tests. A total of 17 fetal hearts were included in our study. In the left ventricle, right ventricle, right atrium, and interventricular septum, the PHH3 index of myocardium was significantly higher over the pericardial region (p-value 0.002, p-value <0.001, <0.001, and 0.009 respectively) as compared to the region of over the endocardium and the middle part of the myocardium. The PHH3 index of the pericardial region of the left ventricle was significantly correlated with the maximum thickness of the left ventricle.
Collapse
Affiliation(s)
- Arundhati Kar
- Anatomy, Institute of Medical Sciences (IMS) and SUM Hospital, Bhubaneswar, IND
| | - Mukund Sable
- Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | - Anbarasan A
- Cytogenetics, Christian Medical College (CMC) Vellore, Vellore, IND
| | - Saubhagya K Jena
- Obstetrics and Gyenacology, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | - Prabhas R Tripathy
- Anatomy, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | - Manisha Gaikwad
- Anatomy, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| |
Collapse
|
15
|
Baghersad S, Sathish Kumar A, Kipper MJ, Popat K, Wang Z. Recent Advances in Tissue-Engineered Cardiac Scaffolds-The Progress and Gap in Mimicking Native Myocardium Mechanical Behaviors. J Funct Biomater 2023; 14:jfb14050269. [PMID: 37233379 DOI: 10.3390/jfb14050269] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Heart failure is the leading cause of death in the US and worldwide. Despite modern therapy, challenges remain to rescue the damaged organ that contains cells with a very low proliferation rate after birth. Developments in tissue engineering and regeneration offer new tools to investigate the pathology of cardiac diseases and develop therapeutic strategies for heart failure patients. Tissue -engineered cardiac scaffolds should be designed to provide structural, biochemical, mechanical, and/or electrical properties similar to native myocardium tissues. This review primarily focuses on the mechanical behaviors of cardiac scaffolds and their significance in cardiac research. Specifically, we summarize the recent development of synthetic (including hydrogel) scaffolds that have achieved various types of mechanical behavior-nonlinear elasticity, anisotropy, and viscoelasticity-all of which are characteristic of the myocardium and heart valves. For each type of mechanical behavior, we review the current fabrication methods to enable the biomimetic mechanical behavior, the advantages and limitations of the existing scaffolds, and how the mechanical environment affects biological responses and/or treatment outcomes for cardiac diseases. Lastly, we discuss the remaining challenges in this field and suggestions for future directions to improve our understanding of mechanical control over cardiac function and inspire better regenerative therapies for myocardial restoration.
Collapse
Affiliation(s)
- Somayeh Baghersad
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Abinaya Sathish Kumar
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Ketul Popat
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
16
|
Bretherton RC, Reichardt IM, Zabrecky KA, Goldstein AJ, Bailey LR, Bugg D, McMillen TS, Kooiker KB, Flint GV, Martinson A, Gunaje J, Koser F, Plaster E, Linke WA, Regnier M, Moussavi-Harami F, Sniadecki NJ, DeForest CA, Davis J. Correcting dilated cardiomyopathy with fibroblast-targeted p38 deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.523684. [PMID: 36747691 PMCID: PMC9900749 DOI: 10.1101/2023.01.23.523684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Inherited mutations in contractile and structural genes, which decrease cardiomyocyte tension generation, are principal drivers of dilated cardiomyopathy (DCM)- the leading cause of heart failure 1,2 . Progress towards developing precision therapeutics for and defining the underlying determinants of DCM has been cardiomyocyte centric with negligible attention directed towards fibroblasts despite their role in regulating the best predictor of DCM severity, cardiac fibrosis 3,4 . Given that failure to reverse fibrosis is a major limitation of both standard of care and first in class precision therapeutics for DCM, this study examined whether cardiac fibroblast-mediated regulation of the heart's material properties is essential for the DCM phenotype. Here we report in a mouse model of inherited DCM that prior to the onset of fibrosis and dilated myocardial remodeling both the myocardium and extracellular matrix (ECM) stiffen from switches in titin isoform expression, enhanced collagen fiber alignment, and expansion of the cardiac fibroblast population, which we blocked by genetically suppressing p38α in cardiac fibroblasts. This fibroblast-targeted intervention unexpectedly improved the primary cardiomyocyte defect in contractile function and reversed ECM and dilated myocardial remodeling. Together these findings challenge the long-standing paradigm that ECM remodeling is a secondary complication to inherited defects in cardiomyocyte contractile function and instead demonstrate cardiac fibroblasts are essential contributors to the DCM phenotype, thus suggesting DCM-specific therapeutics will require fibroblast-specific strategies.
Collapse
|
17
|
Li Y, Zhou Y, Qiao W, Shi J, Qiu X, Dong N. Application of decellularized vascular matrix in small-diameter vascular grafts. Front Bioeng Biotechnol 2023; 10:1081233. [PMID: 36686240 PMCID: PMC9852870 DOI: 10.3389/fbioe.2022.1081233] [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: 10/27/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Coronary artery bypass grafting (CABG) remains the most common procedure used in cardiovascular surgery for the treatment of severe coronary atherosclerotic heart disease. In coronary artery bypass grafting, small-diameter vascular grafts can potentially replace the vessels of the patient. The complete retention of the extracellular matrix, superior biocompatibility, and non-immunogenicity of the decellularized vascular matrix are unique advantages of small-diameter tissue-engineered vascular grafts. However, after vascular implantation, the decellularized vascular matrix is also subject to thrombosis and neoplastic endothelial hyperplasia, the two major problems that hinder its clinical application. The keys to improving the long-term patency of the decellularized matrix as vascular grafts include facilitating early endothelialization and avoiding intravascular thrombosis. This review article sequentially introduces six aspects of the decellularized vascular matrix as follows: design criteria of vascular grafts, components of the decellularized vascular matrix, the changing sources of the decellularized vascular matrix, the advantages and shortcomings of decellularization technologies, modification methods and the commercialization progress as well as the application prospects in small-diameter vascular grafts.
Collapse
Affiliation(s)
| | | | | | | | - Xuefeng Qiu
- *Correspondence: Xuefeng Qiu, ; Nianguo Dong,
| | | |
Collapse
|
18
|
Nelson MS, Liu Y, Wilson HM, Li B, Rosado-Mendez IM, Rogers JD, Block WF, Eliceiri KW. Multiscale Label-Free Imaging of Fibrillar Collagen in the Tumor Microenvironment. Methods Mol Biol 2023; 2614:187-235. [PMID: 36587127 DOI: 10.1007/978-1-0716-2914-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
With recent advances in cancer therapeutics, there is a great need for improved imaging methods for characterizing cancer onset and progression in a quantitative and actionable way. Collagen, the most abundant extracellular matrix protein in the tumor microenvironment (and the body in general), plays a multifaceted role, both hindering and promoting cancer invasion and progression. Collagen deposition can defend the tumor with immunosuppressive effects, while aligned collagen fiber structures can enable tumor cell migration, aiding invasion and metastasis. Given the complex role of collagen fiber organization and topology, imaging has been a tool of choice to characterize these changes on multiple spatial scales, from the organ and tumor scale to cellular and subcellular level. Macroscale density already aids in the detection and diagnosis of solid cancers, but progress is being made to integrate finer microscale features into the process. Here we review imaging modalities ranging from optical methods of second harmonic generation (SHG), polarized light microscopy (PLM), and optical coherence tomography (OCT) to the medical imaging approaches of ultrasound and magnetic resonance imaging (MRI). These methods have enabled scientists and clinicians to better understand the impact collagen structure has on the tumor environment, at both the bulk scale (density) and microscale (fibrillar structure) levels. We focus on imaging methods with the potential to both examine the collagen structure in as natural a state as possible and still be clinically amenable, with an emphasis on label-free strategies, exploiting intrinsic optical properties of collagen fibers.
Collapse
Affiliation(s)
- Michael S Nelson
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuming Liu
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA
| | - Helen M Wilson
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Bin Li
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.,Morgridge Institute for Research, Madison, WI, USA
| | - Ivan M Rosado-Mendez
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeremy D Rogers
- Morgridge Institute for Research, Madison, WI, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Walter F Block
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Kevin W Eliceiri
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA. .,Morgridge Institute for Research, Madison, WI, USA. .,Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA. .,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
19
|
Extracellular Matrix-Based Approaches in Cardiac Regeneration: Challenges and Opportunities. Int J Mol Sci 2022; 23:ijms232415783. [PMID: 36555424 PMCID: PMC9779713 DOI: 10.3390/ijms232415783] [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: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiac development is characterized by the active proliferation of different cardiac cell types, in particular cardiomyocytes and endothelial cells, that eventually build the beating heart. In mammals, these cells lose their regenerative potential early after birth, representing a major obstacle to our current capacity to restore the myocardial structure and function after an injury. Increasing evidence indicates that the cardiac extracellular matrix (ECM) actively regulates and orchestrates the proliferation, differentiation, and migration of cardiac cells within the heart, and that any change in either the composition of the ECM or its mechanical properties ultimately affect the behavior of these cells throughout one's life. Thus, understanding the role of ECMs' proteins and related signaling pathways on cardiac cell proliferation is essential to develop effective strategies fostering the regeneration of a damaged heart. This review provides an overview of the components of the ECM and its mechanical properties, whose function in cardiac regeneration has been elucidated, with a major focus on the strengths and weaknesses of the experimental models so far exploited to demonstrate the actual pro-regenerative capacity of the components of the ECM and to translate this knowledge into new therapies.
Collapse
|
20
|
Watson MC, Williams C, Wang RM, Perreault LR, Sullivan KE, Stoppel WL, Black LD. Extracellular matrix and cyclic stretch alter fetal cardiomyocyte proliferation and maturation in a rodent model of heart hypoplasia. Front Cardiovasc Med 2022; 9:993310. [PMID: 36518682 PMCID: PMC9744115 DOI: 10.3389/fcvm.2022.993310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/10/2022] [Indexed: 01/22/2024] Open
Abstract
Introduction Birth defects, particularly those that affect development of the heart, are a leading cause of morbidity and mortality in infants and young children. Babies born with heart hypoplasia (heart hypoplasia) disorders often have a poor prognosis. It remains unclear whether cardiomyocytes from hypoplastic hearts retain the potential to recover growth, although this knowledge would be beneficial for developing therapies for heart hypoplasia disorders. The objective of this study was to determine the proliferation and maturation potential of cardiomyocytes from hypoplastic hearts and whether these behaviors are influenced by biochemical signaling from the extracellular matrix (ECM) and cyclic mechanical stretch. Method Congenital diaphragmatic hernia (CDH)-associated heart hypoplasia was induced in rat fetuses by maternal exposure to nitrofen. Hearts were isolated from embryonic day 21 nitrofen-treated fetuses positive for CDH (CDH+) and from fetuses without nitrofen administration during gestation. Results and discussion CDH+ hearts were smaller and had decreased myocardial proliferation, along with evidence of decreased maturity compared to healthy hearts. In culture, CDH+ cardiomyocytes remained immature and demonstrated increased proliferative capacity compared to their healthy counterparts. Culture on ECM derived from CDH+ hearts led to a significant reduction in proliferation for both CDH+ and healthy cardiomyocytes. Healthy cardiomyocytes were dosed with exogenous nitrofen to examine whether nitrofen may have an aberrant effect on the proliferative ability of cardiomyocyte, yet no significant change in proliferation was observed. When subjected to stretch, CDH+ cardiomyocytes underwent lengthening of sarcomeres while healthy cardiomyocyte sarcomeres were unaffected. Taken together, our results suggest that alterations to environmental cues such as ECM and stretch may be important factors in the pathological progression of heart hypoplasia.
Collapse
Affiliation(s)
- Matthew C. Watson
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Department of Mechanical Engineering, Tufts University, Medford, MA, United States
| | - Corin Williams
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Raymond M. Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Luke R. Perreault
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Kelly E. Sullivan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Whitney L. Stoppel
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Lauren D. Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
21
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
22
|
Kuwabara JT, Hara A, Heckl JR, Peña B, Bhutada S, DeMaris R, Ivey MJ, DeAngelo LP, Liu X, Park J, Jahansooz JR, Mestroni L, McKinsey TA, Apte SS, Tallquist MD. Regulation of extracellular matrix composition by fibroblasts during perinatal cardiac maturation. J Mol Cell Cardiol 2022; 169:84-95. [PMID: 35569524 PMCID: PMC10149041 DOI: 10.1016/j.yjmcc.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cardiac fibroblasts are the main non-myocyte population responsible for extracellular matrix (ECM) production. During perinatal development, fibroblast expansion coincides with the transition from hyperplastic to hypertrophic myocardial growth. Therefore, we investigated the consequences of fibroblast loss at the time of cardiomyocyte maturation by depleting fibroblasts in the perinatal mouse. METHODS AND RESULTS We evaluated the microenvironment of the perinatal heart in the absence of fibroblasts and the potential functional impact of fibroblast loss in regulation of cardiomyocyte cell cycle arrest and binucleation. Cre-mediated expression of diphtheria toxin A in PDGFRα expressing cells immediately after birth eliminated 70-80% of the cardiac fibroblasts. At postnatal day 5, hearts lacking fibroblasts appeared similar to controls with normal morphology and comparable numbers of endothelial and smooth muscle cells, despite a pronounced reduction in fibrillar collagen. Immunoblotting and proteomic analysis of control and fibroblast-deficient hearts identified differential abundance of several ECM proteins. In addition, fibroblast loss decreased tissue stiffness and resulted in increased cardiomyocyte mitotic index, DNA synthesis, and cytokinesis. Moreover, decellularized matrix from fibroblast-deficient hearts promoted cardiomyocyte DNA replication. While cardiac architecture was not overtly affected by fibroblast reduction, few pups survived past postnatal day 11, suggesting an overall requirement for PDGFRα expressing fibroblasts. CONCLUSIONS These studies demonstrate the key role of fibroblasts in matrix production and cardiomyocyte cross-talk during mouse perinatal heart maturation and revealed that fibroblast-derived ECM may modulate cardiomyocyte maturation in vivo. Neonatal depletion of fibroblasts demonstrated that although hearts can tolerate reduced ECM composition, fibroblast loss eventually leads to perinatal death as the approach simultaneously reduced fibroblast populations in other organs.
Collapse
Affiliation(s)
- Jill T Kuwabara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Akitoshi Hara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Jack R Heckl
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Brisa Peña
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States of America
| | - Regan DeMaris
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Malina J Ivey
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America; Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267, United States of America
| | - Lydia P DeAngelo
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Xiaoting Liu
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Juwon Park
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Julia R Jahansooz
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America
| | - Luisa Mestroni
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States of America
| | - Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States of America.
| |
Collapse
|
23
|
Tan YH, Helms HR, Nakayama KH. Decellularization Strategies for Regenerating Cardiac and Skeletal Muscle Tissues. Front Bioeng Biotechnol 2022; 10:831300. [PMID: 35295645 PMCID: PMC8918733 DOI: 10.3389/fbioe.2022.831300] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is associated with approximately 17.9 million deaths each year. Musculoskeletal conditions affect more than 1.71 billion people globally and are the leading cause of disability. These two areas represent a massive global health burden that is perpetuated by a lack of functionally restorative treatment options. The fields of regenerative medicine and tissue engineering offer great promise for the development of therapies to repair damaged or diseased tissues. Decellularized tissues and extracellular matrices are cornerstones of regenerative biomaterials and have been used clinically for decades and many have received FDA approval. In this review, we first discuss and compare methods used to produce decellularized tissues and ECMs from cardiac and skeletal muscle. We take a focused look at how different biophysical properties such as spatial topography, extracellular matrix composition, and mechanical characteristics influence cell behavior and function in the context of regenerative medicine. Lastly, we describe emerging research and forecast the future high impact applications of decellularized cardiac and skeletal muscle that will drive novel and effective regenerative therapies.
Collapse
Affiliation(s)
| | | | - Karina H. Nakayama
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
24
|
Esmaeili H, Patino-Guerrero A, Hasany M, Ansari MO, Memic A, Dolatshahi-Pirouz A, Nikkhah M. Electroconductive biomaterials for cardiac tissue engineering. Acta Biomater 2022; 139:118-140. [PMID: 34455109 PMCID: PMC8935982 DOI: 10.1016/j.actbio.2021.08.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022]
Abstract
Myocardial infarction (MI) is still the leading cause of mortality worldwide. The success of cell-based therapies and tissue engineering strategies for treatment of injured myocardium have been notably hindered due to the limitations associated with the selection of a proper cell source, lack of engraftment of engineered tissues and biomaterials with the host myocardium, limited vascularity, as well as immaturity of the injected cells. The first-generation approaches in cardiac tissue engineering (cTE) have mainly relied on the use of desired cells (e.g., stem cells) along with non-conductive natural or synthetic biomaterials for in vitro construction and maturation of functional cardiac tissues, followed by testing the efficacy of the engineered tissues in vivo. However, to better recapitulate the native characteristics and conductivity of the cardiac muscle, recent approaches have utilized electroconductive biomaterials or nanomaterial components within engineered cardiac tissues. This review article will cover the recent advancements in the use of electrically conductive biomaterials in cTE. The specific emphasis will be placed on the use of different types of nanomaterials such as gold nanoparticles (GNPs), silicon-derived nanomaterials, carbon-based nanomaterials (CBNs), as well as electroconductive polymers (ECPs) for engineering of functional and electrically conductive cardiac tissues. We will also cover the recent progress in the use of engineered electroconductive tissues for in vivo cardiac regeneration applications. We will discuss the opportunities and challenges of each approach and provide our perspectives on potential avenues for enhanced cTE. STATEMENT OF SIGNIFICANCE: Myocardial infarction (MI) is still the primary cause of death worldwide. Over the past decade, electroconductive biomaterials have increasingly been applied in the field of cardiac tissue engineering. This review article provides the readers with the leading advances in the in vitro applications of electroconductive biomaterials for cTE along with an in-depth discussion of injectable/transplantable electroconductive biomaterials and their delivery methods for in vivo MI treatment. The article also discusses the knowledge gaps in the field and offers possible novel avenues for improved cardiac tissue engineering.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | | | - Masoud Hasany
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | | | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Alireza Dolatshahi-Pirouz
- Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark; Department of Health Technology, Technical University of Denmark, Center for Intestinal Absorption and Transport of Biopharmaceuticals, 2800 Kgs, Lyngby, Denmark
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA; Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
25
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
26
|
Wang X, Pierre V, Senapati S, Park PSH, Senyo SE. Microenvironment Stiffness Amplifies Post-ischemia Heart Regeneration in Response to Exogenous Extracellular Matrix Proteins in Neonatal Mice. Front Cardiovasc Med 2021; 8:773978. [PMID: 34805326 PMCID: PMC8602555 DOI: 10.3389/fcvm.2021.773978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The cardiogenesis of the fetal heart is absent in juveniles and adults. Cross-transplantation of decellularized extracellular matrix (dECM) can stimulate regeneration in myocardial infarct (MI) models. We have previously shown that dECM and tissue stiffness have cooperative regulation of heart regeneration in transiently regenerative day 1 neonatal mice. To investigate underlying mechanisms of mechano-signaling and dECM, we pharmacologically altered heart stiffness and administered dECM hydrogels in non-regenerative mice after MI. The dECM combined with softening exhibits preserved cardiac function, LV geometry, increased cardiomyocyte mitosis and lowered fibrosis while stiffening further aggravated ischemic damage. Transcriptome analysis identified a protein in cardiomyocytes, CLCA2, confirmed to be upregulated after MI and downregulated by dECM in a mechanosensitive manner. Synthetic knock-down of CLCA2 expression induced mitosis in primary rat cardiomyocytes in the dish. Together, our results indicate that therapeutic efficacy of extracellular molecules for heart regeneration can be modulated by heart microenvironment stiffness in vivo.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Valinteshley Pierre
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Paul S.-H. Park
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel E. Senyo
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
27
|
Li S, Ma W, Cai B. Targeting cardiomyocyte proliferation as a key approach of promoting heart repair after injury. MOLECULAR BIOMEDICINE 2021; 2:34. [PMID: 35006441 PMCID: PMC8607366 DOI: 10.1186/s43556-021-00047-y] [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: 01/14/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases such as myocardial infarction (MI) is a major contributor to human mortality and morbidity. The mammalian adult heart almost loses its plasticity to appreciably regenerate new cardiomyocytes after injuries, such as MI and heart failure. The neonatal heart exhibits robust proliferative capacity when exposed to varying forms of myocardial damage. The ability of the neonatal heart to repair the injury and prevent pathological left ventricular remodeling leads to preserved or improved cardiac function. Therefore, promoting cardiomyocyte proliferation after injuries to reinitiate the process of cardiomyocyte regeneration, and suppress heart failure and other serious cardiovascular problems have become the primary goal of many researchers. Here, we review recent studies in this field and summarize the factors that act upon the proliferation of cardiomyocytes and cardiac repair after injury and discuss the new possibilities for potential clinical treatment strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Shuainan Li
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Wenya Ma
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Benzhi Cai
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China. .,Institute of Clinical Pharmacy, the Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, 150086, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, 150086, China.
| |
Collapse
|
28
|
Bahcecioglu G, Yue X, Howe E, Guldner I, Stack MS, Nakshatri H, Zhang S, Zorlutuna P. Aged Breast Extracellular Matrix Drives Mammary Epithelial Cells to an Invasive and Cancer-Like Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100128. [PMID: 34617419 PMCID: PMC8596116 DOI: 10.1002/advs.202100128] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/26/2021] [Indexed: 05/04/2023]
Abstract
Age is a major risk factor for cancer. While the importance of age related genetic alterations in cells on cancer progression is well documented, the effect of aging extracellular matrix (ECM) has been overlooked. This study shows that the aging breast ECM alone is sufficient to drive normal human mammary epithelial cells (KTB21) to a more invasive and cancer-like phenotype, while promoting motility and invasiveness in MDA-MB-231 cells. Decellularized breast matrix from aged mice leads to loss of E-cadherin membrane localization in KTB21 cells, increased cell motility and invasion, and increased production of inflammatory cytokines and cancer-related proteins. The aged matrix upregulates cancer-related genes in KTB21 cells and enriches a cell subpopulation highly expressing epithelial-mesenchymal transition-related genes. Lysyl oxidase knockdown reverts the aged matrix-induced changes to the young levels; it relocalizes E-cadherin to cell membrane, and reduces cell motility, invasion, and cytokine production. These results show for the first time that the aging ECM harbors key biochemical, physical, and mechanical cues contributing to invasive and cancer-like behavior in healthy and cancer mammary cells. Differential response of cells to young and aged ECMs can lead to identification of new targets for cancer treatment and prevention.
Collapse
Affiliation(s)
- Gokhan Bahcecioglu
- Department of Aerospace and Mechanical EngineeringUniversity of Notre DameNotre DameIN46556USA
| | - Xiaoshan Yue
- Department of Aerospace and Mechanical EngineeringUniversity of Notre DameNotre DameIN46556USA
| | - Erin Howe
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIN46556USA
- Department of Biological SciencesUniversity of Notre DameNotre DameIN46556USA
| | - Ian Guldner
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIN46556USA
- Department of Biological SciencesUniversity of Notre DameNotre DameIN46556USA
| | - M. Sharon Stack
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIN46556USA
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIN46556USA
| | - Harikrishna Nakshatri
- Department of SurgerySchool of MedicineIndiana UniversityIndianapolisIN46202USA
- Department of Biochemistry and Molecular BiologySchool of MedicineIndiana UniversityIndianapolisIN46202USA
| | - Siyuan Zhang
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIN46556USA
- Department of Biological SciencesUniversity of Notre DameNotre DameIN46556USA
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical EngineeringUniversity of Notre DameNotre DameIN46556USA
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameIN46556USA
- Bioengineering Graduate ProgramUniversity of Notre DameNotre DameIN46556USA
| |
Collapse
|
29
|
Song MH, Choi SC, Noh JM, Joo HJ, Park CY, Cha JJ, Ahn TH, Ko TH, Choi JI, Na JE, Rhyu IJ, Jang Y, Park Y, Gim JA, Kim JH, Lim DS. LEFTY-PITX2 signaling pathway is critical for generation of mature and ventricular cardiac organoids in human pluripotent stem cell-derived cardiac mesoderm cells. Biomaterials 2021; 278:121133. [PMID: 34571434 DOI: 10.1016/j.biomaterials.2021.121133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.
Collapse
Affiliation(s)
- Myeong-Hwa Song
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea; R&D Center for Companion Diagnostic, SOL Bio Corporation, Suite 510, 27, Seongsui-ro7-gil, Seongdong-gu, Seoul, 04780, South Korea
| | - Ji-Min Noh
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Chi-Yeon Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jung-Joon Cha
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hoon Ahn
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Ji Eun Na
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University Guro Hospital, Seoul,08308, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
30
|
Bongiovanni C, Sacchi F, Da Pra S, Pantano E, Miano C, Morelli MB, D'Uva G. Reawakening the Intrinsic Cardiac Regenerative Potential: Molecular Strategies to Boost Dedifferentiation and Proliferation of Endogenous Cardiomyocytes. Front Cardiovasc Med 2021; 8:750604. [PMID: 34692797 PMCID: PMC8531484 DOI: 10.3389/fcvm.2021.750604] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Despite considerable efforts carried out to develop stem/progenitor cell-based technologies aiming at replacing and restoring the cardiac tissue following severe damages, thus far no strategies based on adult stem cell transplantation have been demonstrated to efficiently generate new cardiac muscle cells. Intriguingly, dedifferentiation, and proliferation of pre-existing cardiomyocytes and not stem cell differentiation represent the preponderant cellular mechanism by which lower vertebrates spontaneously regenerate the injured heart. Mammals can also regenerate their heart up to the early neonatal period, even in this case by activating the proliferation of endogenous cardiomyocytes. However, the mammalian cardiac regenerative potential is dramatically reduced soon after birth, when most cardiomyocytes exit from the cell cycle, undergo further maturation, and continue to grow in size. Although a slow rate of cardiomyocyte turnover has also been documented in adult mammals, both in mice and humans, this is not enough to sustain a robust regenerative process. Nevertheless, these remarkable findings opened the door to a branch of novel regenerative approaches aiming at reactivating the endogenous cardiac regenerative potential by triggering a partial dedifferentiation process and cell cycle re-entry in endogenous cardiomyocytes. Several adaptations from intrauterine to extrauterine life starting at birth and continuing in the immediate neonatal period concur to the loss of the mammalian cardiac regenerative ability. A wide range of systemic and microenvironmental factors or cell-intrinsic molecular players proved to regulate cardiomyocyte proliferation and their manipulation has been explored as a therapeutic strategy to boost cardiac function after injuries. We here review the scientific knowledge gained thus far in this novel and flourishing field of research, elucidating the key biological and molecular mechanisms whose modulation may represent a viable approach for regenerating the human damaged myocardium.
Collapse
Affiliation(s)
- Chiara Bongiovanni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Silvia Da Pra
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Elvira Pantano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Marco Bruno Morelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Gabriele D'Uva
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| |
Collapse
|
31
|
Valussi M, Besser J, Wystub-Lis K, Zukunft S, Richter M, Kubin T, Boettger T, Braun T. Repression of Osmr and Fgfr1 by miR-1/133a prevents cardiomyocyte dedifferentiation and cell cycle entry in the adult heart. SCIENCE ADVANCES 2021; 7:eabi6648. [PMID: 34644107 PMCID: PMC8514096 DOI: 10.1126/sciadv.abi6648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Dedifferentiation of cardiomyocytes is part of the survival program in the remodeling myocardium and may be essential for enabling cardiomyocyte proliferation. In addition to transcriptional processes, non-coding RNAs play important functions for the control of cell cycle regulation in cardiomyocytes and cardiac regeneration. Here, we demonstrate that suppression of FGFR1 and OSMR by miR-1/133a is instrumental to prevent cardiomyocyte dedifferentiation and cell cycle entry in the adult heart. Concomitant inactivation of both miR-1/133a clusters in adult cardiomyocytes activates expression of cell cycle regulators, induces a switch from fatty acid to glycolytic metabolism, and changes expression of extracellular matrix genes. Inhibition of FGFR and OSMR pathways prevents most effects of miR-1/133a inactivation. Short-term miR-1/133a depletion protects cardiomyocytes against ischemia, while extended loss of miR-1/133a causes heart failure. Our results demonstrate a crucial role of miR-1/133a–mediated suppression of Osmr and Ffgfr1 in maintaining the postmitotic differentiated state of cardiomyocytes.
Collapse
Affiliation(s)
- Melissa Valussi
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Johannes Besser
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Katharina Wystub-Lis
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, D-60590 Frankfurt am Main, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, D-61231 Bad Nauheim, Germany
| | - Thomas Kubin
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, D-61231 Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
- Corresponding author. (T.Bo.); (T.Br.)
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Corresponding author. (T.Bo.); (T.Br.)
| |
Collapse
|
32
|
Exogenous extracellular matrix proteins decrease cardiac fibroblast activation in stiffening microenvironment through CAPG. J Mol Cell Cardiol 2021; 159:105-119. [PMID: 34118218 PMCID: PMC10066715 DOI: 10.1016/j.yjmcc.2021.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/20/2022]
Abstract
Controlling fibrosis is an essential part of regenerating the post-ischemic heart. In the post-ischemic heart, fibroblasts differentiate to myofibroblasts that produce collagen-rich matrix to physically stabilize the infarct area. Infarct models in adult mice result in permanent scarring unlike newborn animals which fully regenerate. Decellularized extracellular matrix (dECM) hydrogels derived from early-aged hearts have been shown to be a transplantable therapy that preserves heart function and stimulates cardiomyocyte proliferation and vascularization. In this study, we investigate the anti-fibrotic effects of injectable dECM hydrogels in a cardiac explant model in the context of age-associated tissue compliance. Treatments with adult and fetal dECM hydrogels were tested for molecular effects on cardiac fibroblast activation and fibrosis. Altered sensitivity of fibroblasts to the mechanosignaling of the remodeling microenvironment was evaluated by manipulating the native extracellular matrix in explants and also with elastomeric substrates in the presence of dECM hydrogels. The injectable fetal dECM hydrogel treatment decreases fibroblast activation and contractility and lowers the stiffness-mediated increases in fibroblast activation observed in stiffened explants. The anti-fibrotic effect of dECM hydrogel is most observable at highest stiffness. Experiments with primary cells on elastomeric substrates with dECM treatment support this phenomenon. Transcriptome analysis indicated that dECM hydrogels affect cytoskeleton related signaling including Macrophage capping protein (CAPG) and Leupaxin (LPXN). CAPG was down-regulated by the fetal dECM hydrogel. LPXN expression was decreased by stiffening the explants; however, this effect was reversed by dECM hydrogel treatment. Pharmacological disruption of cytoskeleton polymerization lowered fibroblast activation and CAPG levels. Knocking down CAPG expression with siRNA inhibited fibroblast activation and collagen deposition. Collectively, fibroblast activation is dependent on cooperative action of extracellular molecular signals and mechanosignaling by cytoskeletal integrity.
Collapse
|
33
|
Perreault LR, Le TT, Oudin MJ, Black LD. RNA sequencing indicates age-dependent shifts in the cardiac fibroblast transcriptome between fetal, neonatal, and adult developmental ages. Physiol Genomics 2021; 53:414-429. [PMID: 34281425 PMCID: PMC8560366 DOI: 10.1152/physiolgenomics.00074.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/16/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiac fibroblasts are responsible for extracellular matrix turnover and repair in the cardiac environment and serve to help facilitate immune responses. However, it is well established that they have a significant phenotypic heterogeneity with respect to location, physiological conditions, and developmental age. The goal of this study was to provide an in-depth transcriptomic profile of cardiac fibroblasts derived from rat hearts at fetal, neonatal, and adult developmental ages to ascertain variations in gene expression that may drive functional differences in these cells at these specific stages of development. We performed RNA sequencing (RNA-seq) of cardiac fibroblasts isolated from fetal, neonatal, and adult rats and compared with the rat genome. Principal component analysis of RNA-seq data suggested that data variance was predominantly due to developmental age. Differential expression and gene set enrichment analysis against Gene Ontology and Kyoto Encyclopedia of Genes and Genomes datasets indicated an array of differences across developmental ages, including significant decreases in cardiac development and cardiac function-associated genes with age and a significant increase in immune- and inflammatory-associated functions, particularly immune cell signaling and cytokine and chemokine production, with respect to increasing developmental age. These results reinforce established evidence of diverse phenotypic heterogeneity of fibroblasts with respect to developmental age. Furthermore, based on our analysis of gene expression, age-specific alterations in cardiac fibroblasts may play a crucial role in observed differences in cardiac inflammation and immune response observed across developmental ages.
Collapse
Affiliation(s)
- Luke R Perreault
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Thanh T Le
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
34
|
Park HJ, De Jesus Morales KJ, Bheri S, Kassouf BP, Davis ME. Bidirectional relationship between cardiac extracellular matrix and cardiac cells in ischemic heart disease. Stem Cells 2021; 39:1650-1659. [PMID: 34480804 DOI: 10.1002/stem.3445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/10/2021] [Indexed: 11/07/2022]
Abstract
Ischemic heart diseases (IHDs), including myocardial infarction and cardiomyopathies, are a leading cause of mortality and morbidity worldwide. Cardiac-derived stem and progenitor cells have shown promise as a therapeutic for IHD but are limited by poor cell survival, limited retention, and rapid washout. One mechanism to address this is to encapsulate the cells in a matrix or three-dimensional construct, so as to provide structural support and better mimic the cells' physiological microenvironment during administration. More specifically, the extracellular matrix (ECM), the native cellular support network, has been a strong candidate for this purpose. Moreover, there is a strong consensus that the ECM and its residing cells, including cardiac stem cells, have a constant interplay in response to tissue development, aging, disease progression, and repair. When externally stimulated, the cells and ECM work together to mutually maintain the local homeostasis by initially altering the ECM composition and stiffness, which in turn alters the cellular response and behavior. Given this constant interplay, understanding the mechanism of bidirectional cell-ECM interaction is essential to develop better cell implantation matrices to enhance cell engraftment and cardiac tissue repair. This review summarizes current understanding in the field, elucidating the signaling mechanisms between cardiac ECM and residing cells in response to IHD onset. Furthermore, this review highlights recent advances in native ECM-mimicking cardiac matrices as a platform for modulating cardiac cell behavior and inducing cardiac repair.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Kenneth J De Jesus Morales
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Brandon P Kassouf
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA.,Children's Heart Research and Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
35
|
Ghori FF, Wahid M. Induced pluripotent stem cells derived cardiomyocytes from Duchenne Muscular Dystrophy patients in vitro. Pak J Med Sci 2021; 37:1376-1381. [PMID: 34475915 PMCID: PMC8377888 DOI: 10.12669/pjms.37.5.3104] [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: 06/22/2020] [Revised: 07/27/2020] [Accepted: 04/30/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE This study aimed at the in vitro generation of DMD-cardiomyocytes from patient-specific induced pluripotent stem cells derived from a Pakistani patient for future work on DMD in vitro disease modeling and drug testing for efficacy and toxicity. METHODS This in vitro experimental study was carried out from December 2018 to January 2019 at Stem Cells and Regenerative Medicine Lab (SCRML) at Dow Research Institute of Biotechnology and Biomedical Sciences (DRIBBS), Dow University of Health Sciences (DUHS) Urine derived DMD-iPSCs were used which had been generated previously from a Pakistani DMD patient who had been selected through non-random purposive sampling. These were differentiated towards cardiomyocytes using Cardiomyocytes Differentiation media having specified growth factors and then the molecular characterization of the differentiated cells was done using immunofluorescence. RESULTS Pakistani patient's DMD-Cardiomyocytes were generated and their identity was confirmed by positive immunofluorescence for the expression of cardiac markers NKX2-5 and TNNT-2. CONCLUSION This study aimed for in vitro generation of DMD cardiomyocytes for future application in disease modeling, new drug testing for efficacy and toxicity, as well as for drug-testing for tailored personalized therapy. To the best of our knowledge, this was the first time DMD-Cardiomyocytes were generated from Pakistani DMD patients using their own induced pluripotent stem cells.
Collapse
Affiliation(s)
- Fareeha Faizan Ghori
- Fareeha Faizan Ghori, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Mohsin Wahid
- Mohsin Wahid, Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan. Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
36
|
Floy ME, Givens SE, Matthys OB, Mateyka TD, Kerr CM, Steinberg AB, Silva AC, Zhang J, Mei Y, Ogle BM, McDevitt TC, Kamp TJ, Palecek SP. Developmental lineage of human pluripotent stem cell-derived cardiac fibroblasts affects their functional phenotype. FASEB J 2021; 35:e21799. [PMID: 34339055 PMCID: PMC8349112 DOI: 10.1096/fj.202100523r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/15/2021] [Accepted: 06/30/2021] [Indexed: 01/24/2023]
Abstract
Cardiac fibroblasts (CFBs) support heart function by secreting extracellular matrix (ECM) and paracrine factors, respond to stress associated with injury and disease, and therefore are an increasingly important therapeutic target. We describe how developmental lineage of human pluripotent stem cell-derived CFBs, epicardial (EpiC-FB), and second heart field (SHF-FB) impacts transcriptional and functional properties. Both EpiC-FBs and SHF-FBs exhibited CFB transcriptional programs and improved calcium handling in human pluripotent stem cell-derived cardiac tissues. We identified differences including in composition of ECM synthesized, secretion of growth and differentiation factors, and myofibroblast activation potential, with EpiC-FBs exhibiting higher stress-induced activation potential akin to myofibroblasts and SHF-FBs demonstrating higher calcification and mineralization potential. These phenotypic differences suggest that EpiC-FBs have utility in modeling fibrotic diseases while SHF-FBs are a promising source of cells for regenerative therapies. This work directly contrasts regional and developmental specificity of CFBs and informs CFB in vitro model selection.
Collapse
Affiliation(s)
- Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Sophie E Givens
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Oriane B Matthys
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkley, CA, USA
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Taylor D Mateyka
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Charles M Kerr
- Molecular Cell Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | - Alexandra B Steinberg
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Ana C Silva
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Jianhua Zhang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Ying Mei
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Todd C McDevitt
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
37
|
Gaffney L, Davis Z, Mora-Navarro C, Fisher MB, Freytes DO. Extracellular Matrix Hydrogels Promote Expression of Muscle-Tendon Junction Proteins. Tissue Eng Part A 2021; 28:270-282. [PMID: 34375125 DOI: 10.1089/ten.tea.2021.0070] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Muscle and tendon injuries are prevalent and range from minor sprains and strains to traumatic, debilitating injuries. However, the interactions between these tissues during injury and recovery remain unclear. Three-dimensional tissue models that incorporate both tissues and a physiologically relevant junction between muscle and tendon may help understand how the two tissues interact. Here, we use tissue specific extracellular matrix (ECM) derived from muscle and tendon to determine how cells of each tissue interact with the microenvironment of the opposite tissue resulting in junction specific features. ECM materials were derived from the Achilles tendon and gastrocnemius muscle, decellularized, and processed to form tissue specific pre-hydrogel digests. ECM materials were unique in respect to protein composition and included many types of ECM proteins, not just collagens. After digestion and gelation, ECM hydrogels had similar complex viscosities which were less than type I collagen hydrogels at the same concentration. C2C12 myoblasts and tendon fibroblasts were cultured in tissue-specific ECM conditioned media or encapsulated in tissue-specific ECM hydrogels to determine cell-matrix interactions and the effects on a muscle-tendon junction marker, paxillin. ECM conditioned media had only a minor effect on upregulation of paxillin in cells cultured in monolayer. However, cells cultured within ECM hydrogels had 50-70% higher paxillin expression than cells cultured in type I collagen hydrogels. Contraction of the ECM hydrogels varied by the type of ECM used. Subsequent experiments with varying density of type I collagen (and thus contraction) showed no correlation between paxillin expression and the amount of gel contraction, suggesting that a constituent of the ECM was the driver of paxillin expression in the ECM hydrogels. In addition, the extracellular matrix protein type XXII collagen had similar expression patterns as paxillin, with smaller effect sizes. Using tissue specific ECM allowed for the de-construction of the cell-matrix interactions similar to muscle-tendon junctions to study the expression of MTJ specific proteins.
Collapse
Affiliation(s)
- Lewis Gaffney
- University of North Carolina at Chapel Hill & North Carolina State University, Biomedical Engineering, Raleigh, North Carolina, United States;
| | - Zachary Davis
- University of North Carolina at Chapel Hill & North Carolina State University, Biomedical Engineering, Raleigh, North Carolina, United States;
| | - Camilo Mora-Navarro
- University of North Carolina at Chapel Hill & North Carolina State University, Biomedical Engineering, Raleigh, North Carolina, United States.,North Carolina State University, 6798, Comparative Medicine Institute, Raleigh, North Carolina, United States;
| | - Matthew B Fisher
- University of North Carolina at Chapel Hill & North Carolina State University, Biomedical Engineering, Raleigh, North Carolina, United States.,University of North Carolina at Chapel Hill School of Medicine, 6797, Department of Orthopaedics, Chapel Hill, North Carolina, United States;
| | - Donald O Freytes
- University of North Carolina at Chapel Hill & North Carolina State University, Biomedical Engineering, Raleigh, North Carolina, United States;
| |
Collapse
|
38
|
Batalov I, Jallerat Q, Kim S, Bliley J, Feinberg AW. Engineering aligned human cardiac muscle using developmentally inspired fibronectin micropatterns. Sci Rep 2021; 11:11502. [PMID: 34075068 PMCID: PMC8169656 DOI: 10.1038/s41598-021-87550-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/31/2021] [Indexed: 11/09/2022] Open
Abstract
Cardiac two-dimensional tissues were engineered using biomimetic micropatterns based on the fibronectin-rich extracellular matrix (ECM) of the embryonic heart. The goal of this developmentally-inspired, in vitro approach was to identify cell-cell and cell-ECM interactions in the microenvironment of the early 4-chambered vertebrate heart that drive cardiomyocyte organization and alignment. To test this, biomimetic micropatterns based on confocal imaging of fibronectin in embryonic chick myocardium were created and compared to control micropatterns designed with 2 or 20 µm wide fibronectin lines. Results show that embryonic chick cardiomyocytes have a unique density-dependent alignment on the biomimetic micropattern that is mediated in part by N-cadherin, suggesting that both cell-cell and cell-ECM interactions play an important role in the formation of aligned myocardium. Human induced pluripotent stem cell-derived cardiomyocytes also showed density-dependent alignment on the biomimetic micropattern but were overall less well organized. Interestingly, the addition of human adult cardiac fibroblasts and conditioning with T3 hormone were both shown to increase human cardiomyocyte alignment. In total, these results show that cardiomyocyte maturation state, cardiomyocyte-cardiomyocyte and cardiomyocyte-fibroblast interactions, and cardiomyocyte-ECM interactions can all play a role when engineering anisotropic cardiac tissues in vitro and provides insight as to how these factors may influence cardiogenesis in vivo.
Collapse
Affiliation(s)
- Ivan Batalov
- Department of Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Quentin Jallerat
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Sean Kim
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Jacqueline Bliley
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA
| | - Adam W Feinberg
- Department of Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA. .,Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, 15213, USA.
| |
Collapse
|
39
|
Behmer Hansen RA, Wang X, Kaw G, Pierre V, Senyo SE. Accounting for Material Changes in Decellularized Tissue with Underutilized Methodologies. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6696295. [PMID: 34159202 PMCID: PMC8187050 DOI: 10.1155/2021/6696295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/05/2021] [Accepted: 05/21/2021] [Indexed: 11/17/2022]
Abstract
Tissue decellularization has rapidly developed to be a practical approach in tissue engineering research; biological tissue is cleared of cells resulting in a protein-rich husk as a natural scaffold for growing transplanted cells as a donor organ therapy. Minimally processed, acellular extracellular matrix reproduces natural interactions with cells in vitro and for tissue engineering applications in animal models. There are many decellularization techniques that achieve preservation of molecular profile (proteins and sugars), microstructure features such as organization of ECM layers (interstitial matrix and basement membrane) and organ level macrofeatures (vasculature and tissue compartments). While structural and molecular cues receive attention, mechanical and material properties of decellularized tissues are not often discussed. The effects of decellularization on an organ depend on the tissue properties, clearing mechanism, chemical interactions, solubility, temperature, and treatment duration. Physical characterization by a few labs including work from the authors provides evidence that decellularization protocols should be tailored to specific research questions. Physical characterization beyond histology and immunohistochemistry of the decellularized matrix (dECM) extends evaluation of retained functional features of the original tissue. We direct our attention to current technologies that can be employed for structure function analysis of dECM using underutilized tools such as atomic force microscopy (AFM), cryogenic electron microscopy (cryo-EM), dynamic mechanical analysis (DMA), Fourier-transform infrared spectroscopy (FTIR), mass spectrometry, and rheometry. Structural imaging and mechanical functional testing combined with high-throughput molecular analyses opens a new approach for a deeper appreciation of how cellular behavior is influenced by the isolated microenvironment (specifically dECM). Additionally, the impact of these features with different decellularization techniques and generation of synthetic material scaffolds with desired attributes are informed. Ultimately, this mechanical profiling provides a new dimension to our understanding of decellularized matrix and its role in new applications.
Collapse
Affiliation(s)
- Ryan A. Behmer Hansen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Xinming Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gitanjali Kaw
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Valinteshley Pierre
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Samuel E. Senyo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
40
|
van der Pol A, Bouten CVC. A Brief History in Cardiac Regeneration, and How the Extra Cellular Matrix May Turn the Tide. Front Cardiovasc Med 2021; 8:682342. [PMID: 34095264 PMCID: PMC8172606 DOI: 10.3389/fcvm.2021.682342] [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: 03/18/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Tissue homeostasis is perturbed by stressful events, which can lead to organ dysfunction and failure. This is particularly true for the heart, where injury resulting from myocardial infarction or ischemic heart disease can result in a cascading event ultimately ending with the loss of functional myocardial tissue and heart failure. To help reverse this loss of healthy contractile tissue, researchers have spent decades in the hopes of characterizing a cell source capable of regenerating the injured heart. Unfortunately, these strategies have proven to be ineffective. With the goal of truly understanding cardiac regeneration, researchers have focused on the innate regenerative abilities of zebrafish and neonatal mammals. This has led to the realization that although cells play an important role in the repair of the diseased myocardium, inducing cardiac regeneration may instead lie in the composition of the extra cellular milieu, specifically the extra cellular matrix. In this review we will briefly summarize the current knowledge regarding cell sources used for cardiac regenerative approaches, since these have been extensively reviewed elsewhere. More importantly, by revisiting innate cardiac regeneration observed in zebrafish and neonatal mammals, we will stress the importance the extra cellular matrix has on reactivating this potential in the adult myocardium. Finally, we will address how we can harness the ability of the extra cellular matrix to guide cardiac repair thereby setting the stage of next generation regenerative strategies.
Collapse
Affiliation(s)
- Atze van der Pol
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, Netherlands
| | | |
Collapse
|
41
|
Rafatian N, Vizely K, Al Asafen H, Korolj A, Radisic M. Drawing Inspiration from Developmental Biology for Cardiac Tissue Engineers. Adv Biol (Weinh) 2021; 5:e2000190. [PMID: 34008910 DOI: 10.1002/adbi.202000190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/21/2020] [Indexed: 12/17/2022]
Abstract
A sound understanding of developmental biology is part of the foundation of effective stem cell-derived tissue engineering. Here, the key concepts of cardiac development that are successfully applied in a bioinspired approach to growing engineered cardiac tissues, are reviewed. The native cardiac milieu is studied extensively from embryonic to adult phenotypes, as it provides a resource of factors, mechanisms, and protocols to consider when working toward establishing living tissues in vitro. It begins with the various cell types that constitute the cardiac tissue. It is discussed how myocytes interact with other cell types and their microenvironment and how they change over time from the embryonic to the adult states, with a view on how such changes affect the tissue function and may be used in engineered tissue models. Key embryonic signaling pathways that have been leveraged in the design of culture media and differentiation protocols are presented. The cellular microenvironment, from extracellular matrix chemical and physical properties, to the dynamic mechanical and electrical forces that are exerted on tissues is explored. It is shown that how such microenvironmental factors can inform the design of biomaterials, scaffolds, stimulation bioreactors, and maturation readouts, and suggest considerations for ongoing biomimetic advancement of engineered cardiac tissues and regeneration strategies for the future.
Collapse
Affiliation(s)
- Naimeh Rafatian
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada
| | - Katrina Vizely
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Hadel Al Asafen
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Milica Radisic
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
42
|
Macrophage phenotype and function are dependent upon the composition and biomechanics of the local cardiac tissue microenvironment. Aging (Albany NY) 2021; 13:16938-16956. [PMID: 34292877 PMCID: PMC8312435 DOI: 10.18632/aging.203054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/20/2021] [Indexed: 01/04/2023]
Abstract
Macrophage accumulation and nitrosative stress are known mechanisms underlying age-related cardiovascular pathology and functional decline. The cardiac muscle microenvironment is known to change with age, yet the direct effects of these changes have yet to be studied in-depth. The present study sought to better elucidate the role that biochemical and biomechanical alterations in cardiac tissue have in the altered phenotype and functionality of cardiac resident macrophages observed with increasing age. To accomplish this, naïve bone marrow derived macrophages from young mice were seeded onto either functionalized poly-dimethyl-siloxane hydrogels ranging in stiffness from 2kPA to 64kPA or onto tissue culture plastic, both of which were coated with either young or aged solubilized mouse cardiac extracellular matrix (cECM). Both biomechanical and biochemical alterations were found to have a significant effect on macrophage polarization and function. Increased substrate stiffness was found to promote macrophage morphologies associated with pro-inflammatory macrophage activation, increased expression of pro-inflammatory inducible nitric oxide synthase protein with increased nitric oxide secretion, and attenuated arginase activity and protein expression. Additionally, exposure to aged cECM promoted attenuated responsivity to both canonical pro-inflammatory and anti-inflammatory cytokine signaling cues when compared to young cECM treated cells. These results suggest that both biomechanical and biochemical changes in the cardiovascular system play a role in promoting the age-related shift towards pro-inflammatory macrophage populations associated with cardiovascular disease development.
Collapse
|
43
|
Ly OT, Brown GE, Han YD, Darbar D, Khetani SR. Bioengineering approaches to mature induced pluripotent stem cell-derived atrial cardiomyocytes to model atrial fibrillation. Exp Biol Med (Maywood) 2021; 246:1816-1828. [PMID: 33899540 DOI: 10.1177/15353702211009146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) serve as a robust platform to model several human arrhythmia syndromes including atrial fibrillation (AF). However, the structural, molecular, functional, and electrophysiological parameters of patient-specific iPSC-derived atrial cardiomyocytes (iPSC-aCMs) do not fully recapitulate the mature phenotype of their human adult counterparts. The use of physiologically inspired microenvironmental cues, such as postnatal factors, metabolic conditioning, extracellular matrix (ECM) modulation, electrical and mechanical stimulation, co-culture with non-parenchymal cells, and 3D culture techniques can help mimic natural atrial development and induce a more mature adult phenotype in iPSC-aCMs. Such advances will not only elucidate the underlying pathophysiological mechanisms of AF, but also identify and assess novel mechanism-based therapies towards supporting a more 'personalized' (i.e. patient-specific) approach to pharmacologic therapy of AF.
Collapse
Affiliation(s)
- Olivia T Ly
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yong Duk Han
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA.,Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
44
|
Santos ARMP, Jang Y, Son I, Kim J, Park Y. Recapitulating Cardiac Structure and Function In Vitro from Simple to Complex Engineering. MICROMACHINES 2021; 12:mi12040386. [PMID: 33916254 PMCID: PMC8067203 DOI: 10.3390/mi12040386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiac tissue engineering aims to generate in vivo-like functional tissue for the study of cardiac development, homeostasis, and regeneration. Since the heart is composed of various types of cells and extracellular matrix with a specific microenvironment, the fabrication of cardiac tissue in vitro requires integrating technologies of cardiac cells, biomaterials, fabrication, and computational modeling to model the complexity of heart tissue. Here, we review the recent progress of engineering techniques from simple to complex for fabricating matured cardiac tissue in vitro. Advancements in cardiomyocytes, extracellular matrix, geometry, and computational modeling will be discussed based on a technology perspective and their use for preparation of functional cardiac tissue. Since the heart is a very complex system at multiscale levels, an understanding of each technique and their interactions would be highly beneficial to the development of a fully functional heart in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Jongseong Kim
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| | - Yongdoo Park
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| |
Collapse
|
45
|
Chen W, Bian W, Zhou Y, Zhang J. Cardiac Fibroblasts and Myocardial Regeneration. Front Bioeng Biotechnol 2021; 9:599928. [PMID: 33842440 PMCID: PMC8026894 DOI: 10.3389/fbioe.2021.599928] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
The billions of cardiomyocytes lost to acute myocardial infarction (MI) cannot be replaced by the limited regenerative capacity of adult mammalian hearts, and despite decades of research, there are still no clinically effective therapies for remuscularizing and restoring damaged myocardial tissue. Although the majority of the cardiac mass is composed of cardiomyocytes, cardiac fibroblasts (CFs) are one type of most numerous cells in the heart and the primary drivers of fibrosis, which prevents ventricular rupture immediately after MI but the fibrotic scar expansion and LV dilatation can eventually lead to heart failure. However, embryonic CFs produce cytokines that can activate proliferation in cultured cardiomyocytes, and the structural proteins produced by CFs may regulate cardiomyocyte cell-cycle activity by modulating the stiffness of the extracellular matrix (ECM). CFs can also be used to generate induced-pluripotent stem cells and induced cardiac progenitor cells, both of which can differentiate into cardiomyocytes and vascular cells, but cardiomyocytes appear to be more readily differentiated from iPSCs that have been reprogrammed from CFs than from other cell types. Furthermore, the results from recent studies suggest that cultured CFs, as well as the CFs present in infarcted hearts, can be reprogrammed directly into cardiomyocytes. This finding is very exciting as should we be able to successfully increase the efficiency of this reprogramming, we could remuscularize the injured ventricle and restore the LV function without need the transplantation of cells or cell products. This review summarizes the role of CFs in the innate response to MI and how their phenotypic plasticity and involvement in ECM production might be manipulated to improve cardiac performance in injured hearts.
Collapse
Affiliation(s)
- Wangping Chen
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weihua Bian
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yang Zhou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
46
|
Tsui JH, Leonard A, Camp ND, Long JT, Nawas ZY, Chavanachat R, Smith AST, Choi JS, Dong Z, Ahn EH, Wolf-Yadlin A, Murry CE, Sniadecki NJ, Kim DH. Tunable electroconductive decellularized extracellular matrix hydrogels for engineering human cardiac microphysiological systems. Biomaterials 2021; 272:120764. [PMID: 33798964 DOI: 10.1016/j.biomaterials.2021.120764] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) offer tremendous potential when used to engineer human tissues for drug screening and disease modeling; however, phenotypic immaturity reduces assay reliability when translating in vitro results to clinical studies. To address this, we have developed hybrid hydrogels comprised of decellularized porcine myocardial extracellular matrix (dECM) and reduced graphene oxide (rGO) to provide a more instructive microenvironment for proper cell and tissue development. A tissue-specific protein profile was preserved post-decellularization, and through the modulation of rGO content and degree of reduction, the mechanical and electrical properties of the hydrogels could be tuned. Engineered heart tissues (EHTs) generated using dECM-rGO hydrogel scaffolds and hiPSC-derived cardiomyocytes exhibited significantly increased twitch forces and had increased expression of genes that regulate contractile function. Improvements in various aspects of electrophysiological function, such as calcium-handling, action potential duration, and conduction velocity, were also induced by the hybrid biomaterial. dECM-rGO hydrogels could also be used as a bioink to print cardiac tissues in a high-throughput manner, and these tissues were utilized to assess the proarrhythmic potential of cisapride. Action potential prolongation and beat interval irregularities was observed in dECM-rGO tissues at clinical doses of cisapride, indicating that the enhanced electrophysiological function of these tissues corresponded well with a capability to produce physiologically relevant drug responses.
Collapse
Affiliation(s)
- Jonathan H Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA
| | - Nathan D Camp
- Department of Genome Sciences, University of Washington, Seattle, WA, 98105, USA
| | - Joseph T Long
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | - Zeid Y Nawas
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | | | - Alec S T Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98105, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Charles E Murry
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; Department of Pathology, University of Washington, Seattle, WA, 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA; Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA; Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
47
|
Abstract
The developing heart is formed of two tissue layers separated by an extracellular matrix (ECM) that provides chemical and physical signals to cardiac cells. While deposition of specific ECM components creates matrix diversity, the cardiac ECM is also dynamic, with modification and degradation playing important roles in ECM maturation and function. In this Review, we discuss the spatiotemporal changes in ECM composition during cardiac development that support distinct aspects of heart morphogenesis. We highlight conserved requirements for specific ECM components in human cardiac development, and discuss emerging evidence of a central role for the ECM in promoting heart regeneration.
Collapse
Affiliation(s)
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
48
|
Esmaeili H, Li C, Fu X, Jung JP. Engineering Extracellular Matrix Proteins to Enhance Cardiac Regeneration After Myocardial Infarction. Front Bioeng Biotechnol 2021; 8:611936. [PMID: 33553118 PMCID: PMC7855456 DOI: 10.3389/fbioe.2020.611936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/18/2020] [Indexed: 01/09/2023] Open
Abstract
Engineering microenvironments for accelerated myocardial repair is a challenging goal. Cell therapy has evolved over a few decades to engraft therapeutic cells to replenish lost cardiomyocytes in the left ventricle. However, compelling evidence supports that tailoring specific signals to endogenous cells rather than the direct integration of therapeutic cells could be an attractive strategy for better clinical outcomes. Of many possible routes to instruct endogenous cells, we reviewed recent cases that extracellular matrix (ECM) proteins contribute to enhanced cardiomyocyte proliferation from neonates to adults. In addition, the presence of ECM proteins exerts biophysical regulation in tissue, leading to the control of microenvironments and adaptation for enhanced cardiomyocyte proliferation. Finally, we also summarized recent clinical trials exclusively using ECM proteins, further supporting the notion that engineering ECM proteins would be a critical strategy to enhance myocardial repair without taking any risks or complications of applying therapeutic cardiac cells.
Collapse
Affiliation(s)
- Hamid Esmaeili
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Chaoyang Li
- School of Animal Sciences, Louisiana State University AgCenter, Baton Rouge, LA, United States
| | - Xing Fu
- School of Animal Sciences, Louisiana State University AgCenter, Baton Rouge, LA, United States
| | - Jangwook P Jung
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
49
|
James DS, Campagnola PJ. Recent Advancements in Optical Harmonic Generation Microscopy: Applications and Perspectives. BME FRONTIERS 2021; 2021:3973857. [PMID: 37849910 PMCID: PMC10521653 DOI: 10.34133/2021/3973857] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/14/2020] [Indexed: 10/19/2023] Open
Abstract
Second harmonic generation (SHG) and third harmonic generation (THG) microscopies have emerged as powerful imaging modalities to examine structural properties of a wide range of biological tissues. Although SHG and THG arise from very different contrast mechanisms, the two are complimentary and can often be collected simultaneously using a modified multiphoton microscope. In this review, we discuss the needed instrumentation for these modalities as well as the underlying theoretical principles of SHG and THG in tissue and describe how these can be leveraged to extract unique structural information. We provide an overview of recent advances showing how SHG microscopy has been used to evaluate collagen alterations in the extracellular matrix and how this has been used to advance our knowledge of cancers, fibroses, and the cornea, as well as in tissue engineering applications. Specific examples using polarization-resolved approaches and machine learning algorithms are highlighted. Similarly, we review how THG has enabled developmental biology and skin cancer studies due to its sensitivity to changes in refractive index, which are ubiquitous in all cell and tissue assemblies. Lastly, we offer perspectives and outlooks on future directions of SHG and THG microscopies and present unresolved questions, especially in terms of overall miniaturization and the development of microendoscopy instrumentation.
Collapse
Affiliation(s)
- Darian S. James
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706, USA
| | - Paul J. Campagnola
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706, USA
| |
Collapse
|
50
|
Silva AC, Pereira C, Fonseca ACRG, Pinto-do-Ó P, Nascimento DS. Bearing My Heart: The Role of Extracellular Matrix on Cardiac Development, Homeostasis, and Injury Response. Front Cell Dev Biol 2021; 8:621644. [PMID: 33511134 PMCID: PMC7835513 DOI: 10.3389/fcell.2020.621644] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is an essential component of the heart that imparts fundamental cellular processes during organ development and homeostasis. Most cardiovascular diseases involve severe remodeling of the ECM, culminating in the formation of fibrotic tissue that is deleterious to organ function. Treatment schemes effective at managing fibrosis and promoting physiological ECM repair are not yet in reach. Of note, the composition of the cardiac ECM changes significantly in a short period after birth, concurrent with the loss of the regenerative capacity of the heart. This highlights the importance of understanding ECM composition and function headed for the development of more efficient therapies. In this review, we explore the impact of ECM alterations, throughout heart ontogeny and disease, on cardiac cells and debate available approaches to deeper insights on cell–ECM interactions, toward the design of new regenerative therapies.
Collapse
Affiliation(s)
- Ana Catarina Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Gladstone Institutes, San Francisco, CA, United States
| | - Cassilda Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana Catarina R G Fonseca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Perpétua Pinto-do-Ó
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|