1
|
Coyle A, Chakraborty A, Huang J, Shamiya Y, Luo W, Paul A. In Vitro Engineered ECM-incorporated Hydrogels for Osteochondral Tissue Repair: A Cell-Free Approach. Adv Healthc Mater 2025:e2402701. [PMID: 39757463 DOI: 10.1002/adhm.202402701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/08/2024] [Indexed: 01/07/2025]
Abstract
Prevalence of osteoarthritis has been increasing in aging populations, which has necessitated the use of advanced biomedical treatments. These involve grafts or delivering drug molecules entrapped in scaffolds. However, such treatments often show suboptimal therapeutic effects due to poor half-life and off-target effects of drug molecules. As a countermeasure, a 3D printable robust hydrogel-based tissue-repair platform is developed containing decellularized extracellular matrix (dECM) from differentiated mammalian cells as the therapeutic cargo. Here, pre-osteoblastic and pre-chondrogenic murine cells are differentiated in vitro, decellularized, and incorporated into methacrylated gelatin (GelMA) solutions to form osteogenic (GelO) and chondrogenic (GelC) hydrogels, respectively. Integrating the bioactive dECM from differentiated cell sources allows GelO and GelC to induce differentiation in human adipose-derived stem cells (hASCs) toward osteogenic and chondrogenic lineages. Further, GelO and GelC can be covalently adhered using a carbodiimide coupling reaction, forming a multi-layered hydrogel with potential application as a bioactive osteochondral plug. The designed multi-layered hydrogel can also induce differentiation of hASCs in vitro. In conclusion, the bioactive dECM carrying 3D printed robust hydrogel offers a promising new drug and cell-free therapeutic strategy for bone and cartilage repair and future osteoarthritis management.
Collapse
Affiliation(s)
- Ali Coyle
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Jiaqi Huang
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Wei Luo
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Arghya Paul
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON, N6A 5B9, Canada
- Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada
| |
Collapse
|
2
|
Aglan HA, Ahmed HH, Beherei HH, Abdel-Hady BM, Ekram B, Kishta MS. Generation of cardiomyocytes from stem cells cultured on nanofibrous scaffold: Experimental approach for attenuation of myocardial infarction. Tissue Cell 2024; 89:102461. [PMID: 38991272 DOI: 10.1016/j.tice.2024.102461] [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: 01/31/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
The current study was constructed to fabricate polyamide based nanofibrous scaffolds (NS) and to define the most promising one for the generation of cardiomyocytes from adipose tissue derived mesenchymal stem cells (ADMSCs). This purpose was extended to assess the potentiality of the generated cardiomyocytes in relieving myocardial infarction (MI) in rats. Production and characterization of NSs were carried out. ADMSCs were cultured on NS and induced to differentiate into cardiomyocytes by specific growth factors. Molecular analysis for myocyte-specific enhancer factor 2 C (MEF2C) and alpha sarcomeric actin (α-SCA) expression was done to confirm the differentiation of ADMSCs into cardiomyocytes for further transplantation into MI induced rats. Implantation of cells in MI afflicted rats boosted heart rate, ST height and PR interval and lessened P duration, RR, QTc and QRS intervals. Also, this type of medication minified serum lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) enzymes activity as well as serum and cardiac troponin T (Tn-T) levels and upraised serum and cardiac α-SCA and cardiac connexin 43 (CX 43) levels. Microscopic feature of cardiac tissue sections of rats in the treated groups revealed great renovation in the cardiac microarchitecture. Conclusively, this attempt gains insight into a realistic strategy for recovery of MI through systemic employment of in vitro generated cardiomyocytes.
Collapse
Affiliation(s)
- Hadeer A Aglan
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt.
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials Technology and Mineral Resources Research Institute, National Research Centre, Giza, Egypt
| | - Bothaina M Abdel-Hady
- Polymers and Pigments Department, Chemical Industries Institute, National Research Centre, Giza, Egypt
| | - Basma Ekram
- Polymers and Pigments Department, Chemical Industries Institute, National Research Centre, Giza, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| |
Collapse
|
3
|
Kazemi Asl S, Rahimzadegan M, Ostadrahimi R. The recent advancement in the chitosan hybrid-based scaffolds for cardiac regeneration after myocardial infarction. Carbohydr Polym 2023; 300:120266. [DOI: 10.1016/j.carbpol.2022.120266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/08/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022]
|
4
|
Sakaguchi K, Tobe Y, Yang J, Tanaka RI, Yamanaka K, Ono J, Shimizu T. Bioengineering of a scaffold-less three-dimensional tissue using net mould. Biofabrication 2021; 13. [PMID: 34488209 DOI: 10.1088/1758-5090/ac23e3] [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: 04/16/2021] [Accepted: 09/06/2021] [Indexed: 11/11/2022]
Abstract
Tissue engineering has attracted attention worldwide because of its application in regenerative medicine, drug screening, and cultured meat. Numerous biofabrication techniques for producing tissues have been developed, including various scaffold and printing methods. Here, we have proposed a novel tissue engineering method using a net metal mould without the use of a scaffold. Briefly, normal human dermal fibroblasts seeded on a dimple plate were subjected to static culture technique for several days to form spheroids. Spheroids of diameter ⩾200μm were poured into a net-shaped mould of gap ⩽100μm and subjected to shake-cultivation for several weeks, facilitating their fusion to form a three-dimensional (3D) tissue. Through this study, we successfully constructed a scaffold-free 3D tissue having strength that can be easily manipulated, which was difficult to construct using conventional tissue engineering methods. We also investigated the viability of the 3D tissue and found that the condition of the tissues was completely different depending on the culture media used. Collectively, this method allows scaffold-free culture of 3D tissues of unprecedented thickness, and may contribute largely to next-generation tissue engineering products.
Collapse
Affiliation(s)
- Katsuhisa Sakaguchi
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yusuke Tobe
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Jiayue Yang
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Ryu-Ichiro Tanaka
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kumiko Yamanaka
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Jiro Ono
- TissueByNet Corporation, 24-27-804 Iwafuchi-machi, Kita-ku, Tokyo 115-0041, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
5
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
6
|
Pretorius D, Kahn-Krell AM, Lou X, Fast VG, Berry JL, Kamp TJ, Zhang J. Layer-By-Layer Fabrication of Large and Thick Human Cardiac Muscle Patch Constructs With Superior Electrophysiological Properties. Front Cell Dev Biol 2021; 9:670504. [PMID: 33937272 PMCID: PMC8086556 DOI: 10.3389/fcell.2021.670504] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023] Open
Abstract
Engineered cardiac tissues fabricated from human induced pluripotent stem cells (hiPSCs) show promise for ameliorating damage from myocardial infarction, while also restoring function to the damaged left ventricular (LV) myocardium. For these constructs to reach their clinical potential, they need to be of a clinically relevant volume and thickness, and capable of generating synchronous and forceful contraction to assist the pumping action of the recipient heart. Design prerequisites include a structure thickness sufficient to produce a beneficial contractile force, prevascularization to overcome diffusion limitations and sufficient structural development to allow for maximal cell communication. Previous attempts to meet these prerequisites have been hindered by lack of oxygen and nutrient transport due to diffusion limits (100–200 μm) resulting in necrosis. This study employs a layer-by-layer (LbL) fabrication method to produce cardiac tissue constructs that meet these design prerequisites and mimic normal myocardium in form and function. Thick (>2 mm) cardiac tissues created from hiPSC-derived cardiomyocytes, -endothelial cells (ECs) and -fibroblasts (FBs) were assessed, in vitro, over a 4-week period for viability (<6% necrotic cells), cell morphology and functionality. Functional performance assessment showed enhanced t-tubule network development, gap junction communication as well as previously unseen, physiologically relevant conduction velocities (CVs) (>30 cm/s). These results demonstrate that LbL fabrication can be utilized successfully to create prevascularized, functional cardiac tissue constructs from hiPSCs for potential therapeutic applications.
Collapse
Affiliation(s)
- Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Asher M Kahn-Krell
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xi Lou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vladimir G Fast
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joel L Berry
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 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
|
7
|
Chirico N, Van Laake LW, Sluijter JPG, van Mil A, Dierickx P. Cardiac circadian rhythms in time and space: The future is in 4D. Curr Opin Pharmacol 2020; 57:49-59. [PMID: 33338891 DOI: 10.1016/j.coph.2020.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/25/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
The circadian clock synchronizes the body into 24-h cycles, thereby anticipating variations in tissue-specific diurnal tasks, such as response to increased cardiac metabolic demand during the active period of the day. As a result, blood pressure, heart rate, cardiac output, and occurrence of fatal cardiovascular events fluctuate in a diurnal manner. The heart contains different cell types that make up and reside in an environment of biochemical, mechanical, and topographical signaling. Cardiac architecture is essential for proper heart development as well as for maintenance of cell homeostasis and tissue repair. In this review, we describe the possibilities of studying circadian rhythmicity in the heart by using advanced in vitro systems that mimic the native cardiac 3D microenvironment which can be tuned in time and space. Harnessing the knowledge that originates from those in vitro models could significantly improve innovative cardiac modeling and regenerative strategies.
Collapse
Affiliation(s)
- Nino Chirico
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Linda W Van Laake
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alain van Mil
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Pieterjan Dierickx
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, USA.
| |
Collapse
|
8
|
Pretorius D, Kahn-Krell AM, LaBarge WC, Lou X, Kannappan R, Pollard AE, Fast VG, Berry JL, Eberhardt AW, Zhang J. Fabrication and characterization of a thick, viable bi-layered stem cell-derived surrogate for future myocardial tissue regeneration. Biomed Mater 2020; 16. [PMID: 33053512 DOI: 10.1088/1748-605x/abc107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Cardiac tissue surrogates show promise for restoring mechanical and electrical function in infarcted left ventricular (LV) myocardium. For these cardiac surrogates to be useful in vivo, they are required to support synchronous and forceful contraction over the infarcted region. These design requirements necessitate a thickness sufficient to produce a useful contractile force, an area large enough to cover an infarcted region, and prevascularization to overcome diffusion limitations. Attempts to meet these requirements have been hampered by diffusion limits of oxygen and nutrients (100-200 μm) leading to necrotic regions.This study demonstrates a novel layer-by-layer (LbL) fabrication method used to produce tissue surrogates that meet these requirements and mimic normal myocardium in form and function. Thick (1.5-2 mm) LbL cardiac tissues created from human induced pluripotent stem cell-derived cardiomyocytes and endothelial cells were assessed, in vitro, over a four week period for viability (< 5.6 ± 1.4 % nectrotic cells), cell morphology, viscoelastic properties and functionality. Viscoelastic properties of the cardiac surrogates were determined via stress relaxation response modeling and compared to native murine LV tissue. Viscoelastic characterization showed that the generalized Maxwell model of order 4 described the samples well (0.7 < R2 < 0.98). Functional performance assessment showed enhanced t-tubule network development, gap junction communication as well as conduction velocity (16.9 ± 2.3 cm s-1). These results demonstrate that LbL fabrication can be utilized successfully in creating complex, functional cardiac surrogates for therapeutic applications.
Collapse
Affiliation(s)
- Danielle Pretorius
- Biomedical Engineering, The University of Alabama at Birmingham, Volker Hall Room G094, 1670 University Blvd, Birmingham, Alabama, 35294-2182, UNITED STATES
| | - Asher M Kahn-Krell
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Wesley C LaBarge
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Xi Lou
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Ramaswamy Kannappan
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Andrew E Pollard
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Vladimir G Fast
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Joel L Berry
- School of Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Alan W Eberhardt
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Jianyi Zhang
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| |
Collapse
|
9
|
Theus AS, Ning L, Hwang B, Gil C, Chen S, Wombwell A, Mehta R, Serpooshan V. Bioprintability: Physiomechanical and Biological Requirements of Materials for 3D Bioprinting Processes. Polymers (Basel) 2020; 12:E2262. [PMID: 33019639 PMCID: PMC7599870 DOI: 10.3390/polym12102262] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
Three-dimensional (3D) bioprinting is an additive manufacturing process that utilizes various biomaterials that either contain or interact with living cells and biological systems with the goal of fabricating functional tissue or organ mimics, which will be referred to as bioinks. These bioinks are typically hydrogel-based hybrid systems with many specific features and requirements. The characterizing and fine tuning of bioink properties before, during, and after printing are therefore essential in developing reproducible and stable bioprinted constructs. To date, myriad computational methods, mechanical testing, and rheological evaluations have been used to predict, measure, and optimize bioinks properties and their printability, but none are properly standardized. There is a lack of robust universal guidelines in the field for the evaluation and quantification of bioprintability. In this review, we introduced the concept of bioprintability and discussed the significant roles of various physiomechanical and biological processes in bioprinting fidelity. Furthermore, different quantitative and qualitative methodologies used to assess bioprintability will be reviewed, with a focus on the processes related to pre, during, and post printing. Establishing fully characterized, functional bioink solutions would be a big step towards the effective clinical applications of bioprinted products.
Collapse
Affiliation(s)
- Andrea S. Theus
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Carmen Gil
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Shuai Chen
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Allison Wombwell
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
| | - Riya Mehta
- Department of Biology, Emory University, Atlanta, GA 30322, USA;
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (B.H.); (C.G.); (S.C.); (A.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Du Y, Zhang S, Yu H, Wu Y, Cao N, Wang W, Xu W, Li Y, Liu H. Autoantibodies Against β 1-Adrenoceptor Exaggerated Ventricular Remodeling by Inhibiting CTRP9 Expression. J Am Heart Assoc 2020; 8:e010475. [PMID: 30764693 PMCID: PMC6405676 DOI: 10.1161/jaha.118.010475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Autoantibodies against the second extracellular loop of the β1‐adrenoceptor (β1‐AA) act similarly to agonist of β1‐adrenergic receptor, which plays an important role in the pathophysiological characteristics of ventricular remodeling. Recently, considerable lines of evidence have suggested that CTRP9 (C1q tumor necrosis factor–related protein 9) is a potent cardioprotective cardiokine and protects the heart from ventricular remodeling. The aim of this study was to determine the role of CTRP9 in ventricular remodeling induced by β1‐AA. Methods and Results Blood samples were collected from 131 patients with coronary heart disease and 131 healthy subjects. The serum levels of β1‐AA and CTRP9 were detected using ELISA. The results revealed that CTRP9 levels in β1‐AA–positive patients were lower than those in β1‐AA–negative patients, and serum CTRP9 concentrations were inversely correlated with β1‐AA. β1‐AA monoclonal antibodies (β1‐AAmAbs) were administered in mice with and without rAAV9‐cTnT‐Full Ctrp9‐FLAG virus for 8 weeks. Reverse transcription–polymerase chain reaction/Western analysis showed that cardiomyocyte CTRP9 expression was significantly reduced in β1‐AAmAb–treated mice. Moreover, compared with the β1‐AAmAb alone group, cardiac‐specific CTRP9 overexpression improved cardiac function, attenuated adverse remodeling, and ameliorated cardiomyocyte apoptosis and fibrosis. Mechanistic studies demonstrated that CTRP9 overexpression decreased the levels of G‐protein–coupled receptor kinase 2 and promoted the activation of AMP‐dependent kinase pathway. However, cardiac‐specific overexpression of CTRP9 had no effect on the levels of cAMP and protein kinase A activity elevated by β1‐AAmAb. Conclusions This study provides the first evidence that the long‐term existence of β1‐AAmAb suppresses cardiac CTRP9 expression and exaggerates cardiac remodeling, suggesting that CTRP9 may be a novel therapeutic target against pathologic remodeling in β1‐AA–positive patients with coronary heart disease.
Collapse
Affiliation(s)
- Yunhui Du
- 1 Beijing Anzhen Hospital Capital Medical University Beijing Institute of Heart, Lung and Blood Vessel Diseases Beijing China
| | - Shihan Zhang
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Haicun Yu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China.,4 Zhengzhou Central Hospital affiliated of Zhengzhou University Henan Province China
| | - Ye Wu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Ning Cao
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Wen Wang
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Wenli Xu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Yuming Li
- 3 Department of Basic Medical Sciences Yanjing Medical College Capital Medical University Beijing China
| | - Huirong Liu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| |
Collapse
|
11
|
Identification of a pro-angiogenic functional role for FSP1-positive fibroblast subtype in wound healing. Nat Commun 2019; 10:3027. [PMID: 31289275 PMCID: PMC6617456 DOI: 10.1038/s41467-019-10965-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fibrosis accompanying wound healing can drive the failure of many different organs. Activated fibroblasts are the principal determinants of post-injury pathological fibrosis along with physiological repair, making them a difficult therapeutic target. Although activated fibroblasts are phenotypically heterogeneous, they are not recognized as distinct functional entities. Using mice that express GFP under the FSP1 or αSMA promoter, we characterized two non-overlapping fibroblast subtypes from mouse hearts after myocardial infarction. Here, we report the identification of FSP1-GFP+ cells as a non-pericyte, non-hematopoietic fibroblast subpopulation with a predominant pro-angiogenic role, characterized by in vitro phenotypic/cellular/ultrastructural studies and in vivo granulation tissue formation assays combined with transcriptomics and proteomics. This work identifies a fibroblast subtype that is functionally distinct from the pro-fibrotic αSMA-expressing myofibroblast subtype. Our study has the potential to shift our focus towards viewing fibroblasts as molecularly and functionally heterogeneous and provides a paradigm to approach treatment for organ fibrosis. Activated fibroblasts are key contributors to tissue repair after cardiac injury. Here, Saraswati et al. identify and characterize a subpopulation of FSP1-positive cardiac fibroblasts with proangiogenic properties in infarcted hearts.
Collapse
|
12
|
Therapeutic Cell Protective Role of Histochrome under Oxidative Stress in Human Cardiac Progenitor Cells. Mar Drugs 2019; 17:md17060368. [PMID: 31234277 PMCID: PMC6628112 DOI: 10.3390/md17060368] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiac progenitor cells (CPCs) are resident stem cells present in a small portion of ischemic hearts and function in repairing the damaged heart tissue. Intense oxidative stress impairs cell metabolism thereby decreasing cell viability. Protecting CPCs from undergoing cellular apoptosis during oxidative stress is crucial in optimizing CPC-based therapy. Histochrome (sodium salt of echinochrome A—a common sea urchin pigment) is an antioxidant drug that has been clinically used as a pharmacologic agent for ischemia/reperfusion injury in Russia. However, the mechanistic effect of histochrome on CPCs has never been reported. We investigated the protective effect of histochrome pretreatment on human CPCs (hCPCs) against hydrogen peroxide (H2O2)-induced oxidative stress. Annexin V/7-aminoactinomycin D (7-AAD) assay revealed that histochrome-treated CPCs showed significant protective effects against H2O2-induced cell death. The anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and Bcl-xL were significantly upregulated, whereas the pro-apoptotic proteins BCL2-associated X (Bax), H2O2-induced cleaved caspase-3, and the DNA damage marker, phosphorylated histone (γH2A.X) foci, were significantly downregulated upon histochrome treatment of hCPCs in vitro. Further, prolonged incubation with histochrome alleviated the replicative cellular senescence of hCPCs. In conclusion, we report the protective effect of histochrome against oxidative stress and present the use of a potent and bio-safe cell priming agent as a potential therapeutic strategy in patient-derived hCPCs to treat heart disease.
Collapse
|
13
|
Bax NAM, Duim SN, Kruithof BPT, Smits AM, Bouten CVC, Goumans MJ. In vivo and in vitro Approaches Reveal Novel Insight Into the Ability of Epicardium-Derived Cells to Create Their Own Extracellular Environment. Front Cardiovasc Med 2019; 6:81. [PMID: 31275946 PMCID: PMC6594358 DOI: 10.3389/fcvm.2019.00081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Human epicardium-derived cells (hEPDCs) transplanted in the NOD-SCID mouse heart after myocardial infarction (MI) are known to improve cardiac function, most likely orchestrated by paracrine mechanisms that limit adverse remodeling. It is not yet known, however, if hEPDCs contribute to preservation of cardiac function via the secretion of matrix proteins and/or matrix proteases to reduce scar formation. This study describes the ability of hEPDCs to produce human collagen type I after transplantation into the infarct border zone, thereby creating their own extracellular environment. As the in vivo environment is too complex to investigate the mechanisms involved, we use an in vitro set-up, mimicking biophysical and biochemical cues from the myocardial tissue to unravel hEPDC-induced matrix remodeling. The in vivo contribution of hEPDCs to the cardiac extracellular matrix (ECM) was assessed in a historical dataset of the NOD-SCID murine model of experimentally induced MI and cell transplantation. Analysis showed that within 48 h after transplantation, hEPDCs produce human collagen type I. The build-up of the human collagen microenvironment was reversed within 6 weeks. To understand the hEPDCs response to the pathologic cardiac microenvironment, we studied the influence of cyclic straining and/or transforming growth beta (TGFβ) signaling in vitro. We revealed that 48 h of cyclic straining induced collagen type I production via the TGFβ/ALK5 signaling pathway. The in vitro approach enables further unraveling of the hEPDCs ability to secrete matrix proteins and matrix proteases and the potential to create and remodel the cardiac matrix in response to injury.
Collapse
Affiliation(s)
- Noortje A M Bax
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sjoerd N Duim
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Boudewijn P T Kruithof
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Anke M Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Marie José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
14
|
Spaans S, Fransen PPKH, Schotman MJG, van der Wulp R, Lafleur RP, Kluijtmans SGJM, Dankers PYW. Supramolecular Modification of a Sequence-Controlled Collagen-Mimicking Polymer. Biomacromolecules 2019; 20:2360-2371. [PMID: 31050892 PMCID: PMC6560502 DOI: 10.1021/acs.biomac.9b00353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/29/2019] [Indexed: 11/29/2022]
Abstract
Structurally and functionally well-defined recombinant proteins are an interesting class of sequence-controlled macromolecules to which different crosslinking chemistries can be applied to tune their biological properties. Herein, we take advantage of a 571-residue recombinant peptide based on human collagen type I (RCPhC1), which we functionalized with supramolecular 4-fold hydrogen bonding ureido-pyrimidinone (UPy) moieties. By grafting supramolecular UPy moieties onto the backbone of RCPhC1 (UPy-RCPhC1), increased control over the polymer structure, assembly, gelation, and mechanical properties was achieved. In addition, by increasing the degree of UPy functionalization on RCPhC1, cardiomyocyte progenitor cells were cultured on "soft" (∼26 kPa) versus "stiff" (∼68-190 kPa) UPy-RCPhC1 hydrogels. Interestingly, increased stress fiber formation, focal adhesions, and proliferation were observed on stiffer compared to softer substrates, owing to the formation of stronger cell-material interactions. In conclusion, a bioinspired hydrogel material was designed by a combination of two well-known natural components, i.e., a protein as sequence-controlled polymer and UPy units inspired on nucleobases.
Collapse
Affiliation(s)
- Sergio Spaans
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Peter-Paul K. H. Fransen
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Maaike J. G. Schotman
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ruben van der Wulp
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - René P.
M. Lafleur
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
15
|
Dunn KK, Palecek SP. Engineering Scalable Manufacturing of High-Quality Stem Cell-Derived Cardiomyocytes for Cardiac Tissue Repair. Front Med (Lausanne) 2018; 5:110. [PMID: 29740580 PMCID: PMC5928319 DOI: 10.3389/fmed.2018.00110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/03/2018] [Indexed: 12/29/2022] Open
Abstract
Recent advances in the differentiation and production of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) have stimulated development of strategies to use these cells in human cardiac regenerative therapies. A prerequisite for clinical trials and translational implementation of hPSC-derived CMs is the ability to manufacture safe and potent cells on the scale needed to replace cells lost during heart disease. Current differentiation protocols generate fetal-like CMs that exhibit proarrhythmogenic potential. Sufficient maturation of these hPSC-derived CMs has yet to be achieved to allow these cells to be used as a regenerative medicine therapy. Insights into the native cardiac environment during heart development may enable engineering of strategies that guide hPSC-derived CMs to mature. Specifically, considerations must be made in regard to developing methods to incorporate the native intercellular interactions and biomechanical cues into hPSC-derived CM production that are conducive to scale-up.
Collapse
Affiliation(s)
- Kaitlin K Dunn
- University of Wisconsin-Madison, Chemical and Biological Engineering, Madison, WI, United States
| | - Sean P Palecek
- University of Wisconsin-Madison, Chemical and Biological Engineering, Madison, WI, United States
| |
Collapse
|
16
|
Spaans S, Fransen PPKH, Ippel BD, de Bont DFA, Keizer HM, Bax NAM, Bouten CVC, Dankers PYW. Supramolecular surface functionalization via catechols for the improvement of cell-material interactions. Biomater Sci 2018. [PMID: 28636048 DOI: 10.1039/c7bm00407a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Optimization of cell-material interactions is crucial for the success of synthetic biomaterials in guiding tissue regeneration. To do so, catechol chemistry is often used to introduce adhesiveness into biomaterials. Here, a supramolecular approach based on ureido-pyrimidinone (UPy) modified polymers is combined with catechol chemistry in order to achieve improved cellular adhesion onto supramolecular biomaterials. UPy-modified hydrophobic polymers with non-cell adhesive properties are developed that can be bioactivated via a modular approach using UPy-modified catechols. It is shown that successful formulation of the UPy-catechol additive with the UPy-polymer results in surfaces that induce cardiomyocyte progenitor cell adhesion, cell spreading, and preservation of cardiac specific extracellular matrix production. Hence, by functionalizing supramolecular surfaces with catechol functionalities, an adhesive supramolecular biomaterial is developed that allows for the possibility to contribute to biomaterial-based regeneration.
Collapse
Affiliation(s)
- S Spaans
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - P P K H Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. box 513, 5600 MB Eindhoven, The Netherlands
| | - B D Ippel
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - D F A de Bont
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - H M Keizer
- SyMO-Chem BV, Eindhoven University of Technology, De Zaale, 5612 AZ Eindhoven, The Netherlands
| | - N A M Bax
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - C V C Bouten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - P Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands and Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
17
|
Cardiac Progenitor Cells and the Interplay with Their Microenvironment. Stem Cells Int 2017; 2017:7471582. [PMID: 29075298 PMCID: PMC5623801 DOI: 10.1155/2017/7471582] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells, extracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity.
Collapse
|
18
|
Lee S, Serpooshan V, Tong X, Venkatraman S, Lee M, Lee J, Chirikian O, Wu JC, Wu SM, Yang F. Contractile force generation by 3D hiPSC-derived cardiac tissues is enhanced by rapid establishment of cellular interconnection in matrix with muscle-mimicking stiffness. Biomaterials 2017; 131:111-120. [PMID: 28384492 DOI: 10.1016/j.biomaterials.2017.03.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 02/07/2023]
Abstract
Engineering 3D human cardiac tissues is of great importance for therapeutic and pharmaceutical applications. As cardiac tissue substitutes, extracellular matrix-derived hydrogels have been widely explored. However, they exhibit premature degradation and their stiffness is often orders of magnitude lower than that of native cardiac tissue. There are no reports on establishing interconnected cardiomyocytes in 3D hydrogels at physiologically-relevant cell density and matrix stiffness. Here we bioengineer human cardiac microtissues by encapsulating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in chemically-crosslinked gelatin hydrogels (1.25 × 108/mL) with tunable stiffness and degradation. In comparison to the cells in high stiffness (16 kPa)/slow degrading hydrogels, hiPSC-CMs in low stiffness (2 kPa)/fast degrading and intermediate stiffness (9 kPa)/intermediate degrading hydrogels exhibit increased intercellular network formation, α-actinin and connexin-43 expression, and contraction velocity. Only the 9 kPa microtissues exhibit organized sarcomeric structure and significantly increased contractile stress. This demonstrates that muscle-mimicking stiffness together with robust cellular interconnection contributes to enhancement in sarcomeric organization and contractile function of the engineered cardiac tissue. This study highlights the importance of intercellular connectivity, physiologically-relevant cell density, and matrix stiffness to best support 3D cardiac tissue engineering.
Collapse
Affiliation(s)
- Soah Lee
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Vahid Serpooshan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Xinming Tong
- Department of Orthopedic Surgery, Stanford, CA, USA
| | - Sneha Venkatraman
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Biology Program, California State University Channel Islands, Camarillo, CA, USA
| | - Meelim Lee
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jaecheol Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Orlando Chirikian
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Biology Program, California State University Channel Islands, Camarillo, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA.
| | - Fan Yang
- Department of Orthopedic Surgery, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Bildyug N. Matrix metalloproteinases: an emerging role in regulation of actin microfilament system. Biomol Concepts 2017; 7:321-329. [PMID: 27763882 DOI: 10.1515/bmc-2016-0022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in many physiological and pathological processes, including contraction, migration, differentiation, and proliferation. These processes all involve cell phenotype changes, known to be accompanied by reorganization of actin cytoskeleton. Growing evidence indicates a correlation between MMP activity and the dynamics of actin system, suggesting their mutual regulation. Here, data on the influence of MMPs on the actin microfilament system, on the one hand, and the dependence of MMP expression and activation on the organization of actin structures, on the other hand, are reviewed. The different mechanisms of putative actin-MMP regulation are discussed.
Collapse
|
20
|
Barabadi Z, Azami M, Sharifi E, Karimi R, Lotfibakhshaiesh N, Roozafzoon R, Joghataei MT, Ai J. Fabrication of hydrogel based nanocomposite scaffold containing bioactive glass nanoparticles for myocardial tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1137-46. [DOI: 10.1016/j.msec.2016.08.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 01/29/2023]
|
21
|
Bildyug NB, Voronkina IV, Smagina LV, Yudintseva NM, Pinaev GP. Matrix Metalloproteinases in Primary Culture of Cardiomyocytes. BIOCHEMISTRY (MOSCOW) 2016; 80:1318-26. [PMID: 26567576 DOI: 10.1134/s0006297915100132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The highly organized contractile apparatus of cardiomyocytes in heart tissue allows for their continuous contractility, whereas extracellular matrix components are synthesized and spatially organized by fibroblasts and endothelial cells. However, reorganization of the cardiomyocyte contractile apparatus occurs upon their 2D cultivation, which is accompanied by transient loss of their contractility and acquired capability of extracellular matrix synthesis (Bildyug, N. B., and Pinaev, G. P. (2013) Tsitologiya, 55, 713-724). In this study, matrix metalloproteinases were investigated at different times of cardiomyocyte 2D cultivation and 3D cultivation in collagen gels. It was found that cardiomyocytes in 2D culture synthesize matrix metalloproteinases MMP-2 and MMP-9, wherein their amount varies with the cultivation time. The peak MMP-9 amount is at early cultivation time, when the reorganization of cardiomyocyte contractile apparatus occurs, and the MMP-2 peak precedes the recovery of the initial organization of their contractile apparatus. Upon cardiomyocyte cultivation in 3D collagen gels, in which case their contractile apparatus does not rearrange, a steady small amount of MMP-2 and MMP-9 is observed. These data indicate that the cardiomyocyte contractile apparatus reorganization in culture is associated with synthesis and spatial organization of their own extracellular matrix.
Collapse
Affiliation(s)
- N B Bildyug
- Institute of Cytology, Russian Academy of Sciences, Russia.
| | | | | | | | | |
Collapse
|
22
|
Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat Commun 2016; 7:12260. [PMID: 27447449 PMCID: PMC5512625 DOI: 10.1038/ncomms12260] [Citation(s) in RCA: 635] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/15/2016] [Indexed: 01/18/2023] Open
Abstract
Cardiac fibroblasts convert to myofibroblasts with injury to mediate healing after acute myocardial infarction (MI) and to mediate long-standing fibrosis with chronic disease. Myofibroblasts remain a poorly defined cell type in terms of their origins and functional effects in vivo. Here we generate Postn (periostin) gene-targeted mice containing a tamoxifen-inducible Cre for cellular lineage-tracing analysis. This Postn allele identifies essentially all myofibroblasts within the heart and multiple other tissues. Lineage tracing with four additional Cre-expressing mouse lines shows that periostin-expressing myofibroblasts in the heart derive from tissue-resident fibroblasts of the Tcf21 lineage, but not endothelial, immune/myeloid or smooth muscle cells. Deletion of periostin+ myofibroblasts reduces collagen production and scar formation after MI. Periostin-traced myofibroblasts also revert back to a less-activated state upon injury resolution. Our results define the myofibroblast as a periostin-expressing cell type necessary for adaptive healing and fibrosis in the heart, which arises from Tcf21+ tissue-resident fibroblasts. The origin and fate of myofibroblasts, the cells responsible for cardiac remodelling and fibrosis, is controversial. Here the authors show that cardiac myofibroblasts express periostin, derive exclusively from tissue-resident fibroblasts, are necessary for scar formation after injury, and can revert back to a less-activated state upon injury resolution.
Collapse
|
23
|
Kapeller B, Mueller J, Losert U, Podesser BK, Macfelda K. Microcurrent stimulation promotes reverse remodelling in cardiomyocytes. ESC Heart Fail 2016; 3:122-130. [PMID: 27774272 PMCID: PMC5064659 DOI: 10.1002/ehf2.12080] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 09/28/2015] [Accepted: 10/27/2015] [Indexed: 01/18/2023] Open
Abstract
Aims It has been shown that electrical stimulation can improve tissue repair in patients. Imbalances in the extracellular matrix composition induce manifestation of heart failure. Here we investigated the application of microcurrent (MC) to modulate the expression of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) in cardiomyocytes in vitro and in vivo to reverse remodelling in the heart in spontaneous hypertensive rats (SHR). Methods Cardiomyocytes from young SHR (7 months) and old SHR (14 months) were stimulated in vitro and in vivo with MC. MMP and TIMP expression were analysed by qPCR and immunofluorescence to evaluate the modulation of MC treatment. Results Modulation of cardiomyocytes with MC enhances proliferation with no morphological changes in vitro. By electrical stimulation dual effects, increase and decrease, on MMP‐2, MMP‐9, TIMP‐3, and TIMP‐4 mRNA as well as protein expression were observed, depending on the age of the cardiomyocytes. In our in vivo study, MC down‐regulated MMP‐2, MMP‐9, and TIMP‐4 and increased TIMP‐3 in young SHR. In old SHR MMP‐2, MMP‐9, and TIMP‐4 were up‐regulated, whereas TIMP‐3 was unaffected. Conclusions Our data indicate that treatment of MC can modulate the expression of MMPs and TIMPs in vitro and in vivo in SHR. Based on these results new treatments for heart failure could be developed.
Collapse
Affiliation(s)
- Barbara Kapeller
- Department for Biomedical Research Medical University Vienna Vienna Austria
| | | | - Udo Losert
- Department for Biomedical Research Medical University Vienna Vienna Austria
| | - Bruno K Podesser
- Department for Biomedical Research Medical University Vienna Vienna Austria
| | - Karin Macfelda
- Department for Biomedical Research Medical University Vienna Vienna Austria
| |
Collapse
|
24
|
Mauretti A, Bax NAM, van Marion MH, Goumans MJ, Sahlgren C, Bouten CVC. Cardiomyocyte progenitor cell mechanoresponse unrevealed: strain avoidance and mechanosome development. Integr Biol (Camb) 2016; 8:991-1001. [DOI: 10.1039/c6ib00117c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The mechanosensitivity of cardiomyocyte progenitor cells (CMPCs) is developed upon early cardiac differentiation, together with the development of the mechanosome.
Collapse
Affiliation(s)
- Arianna Mauretti
- Department of Biomedical Engineering
- Eindhoven University of Technology
- 5600 MB Eindhoven
- The Netherlands
- Institute for Complex Molecular Systems
| | - Noortje A. M. Bax
- Department of Biomedical Engineering
- Eindhoven University of Technology
- 5600 MB Eindhoven
- The Netherlands
- Institute for Complex Molecular Systems
| | - Mieke H. van Marion
- Department of Biomedical Engineering
- Eindhoven University of Technology
- 5600 MB Eindhoven
- The Netherlands
- Institute for Complex Molecular Systems
| | - Marie José Goumans
- Department of Molecular Cell Biology and Center for Biomedical Genetics
- Leiden University Medical Center
- The Netherlands
| | - Cecilia Sahlgren
- Department of Biomedical Engineering
- Eindhoven University of Technology
- 5600 MB Eindhoven
- The Netherlands
- Institute for Complex Molecular Systems
| | - Carlijn V. C. Bouten
- Department of Biomedical Engineering
- Eindhoven University of Technology
- 5600 MB Eindhoven
- The Netherlands
- Institute for Complex Molecular Systems
| |
Collapse
|
25
|
van Marion MH, Bax NA, van Turnhout M, Mauretti A, van der Schaft DW, Goumans MJT, Bouten CV. Behavior of CMPCs in unidirectional constrained and stress-free 3D hydrogels. J Mol Cell Cardiol 2015; 87:79-91. [DOI: 10.1016/j.yjmcc.2015.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/03/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
|
26
|
Li CC, Yao HT, Cheng FJ, Hsieh YH, Lu CY, Wu CC, Liu KL, Chang JW. Docosahexaenoic Acid Downregulates EGF-Induced Urokinase Plasminogen Activator and Matrix Metalloproteinase 9 Expression by Inactivating EGFR/ErbB2 Signaling in SK-BR3 Breast Cancer Cells. Nutr Cancer 2015; 67:771-82. [PMID: 25970488 DOI: 10.1080/01635581.2015.1037961] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Urokinase plasminogen activator (uPA) and matrix metalloproteinase 9 (MMP-9) play crucial roles in tumor metastasis. Despite the well-known anticancer role of docosa-hexaenoic acid (DHA), its specific effect on ErbB2-mediated breast cancer metastasis is not fully clarified. In this study, we investigated the effect of DHA on epidermal growth factor (EGF)-induced uPA and MMP-9 activity, expression and cell invasion in SK-BR3 breast cancer cells and the possible mechanisms involved. The results showed that EGF (40 ng/ml) induced uPA and MMP-9 mRNA and protein expression, enzyme activity, and 100 μM DHA significantly inhibited EGF-induced uPA and MMP-9 mRNA, protein expression, enzyme activity, cell migration, and cell invasion. EGF increased protein expression and phosphorylation of EGF receptor (EGFR) and ErbB2 as well as of JNK2, ERK1/2, and Akt, and these changes were attenuated by DHA pretreatment. AG1478, an inhibitor of EGFR, also attenuated EGF-induced activation of EGFR, JNK2, ERK1/2, and Akt. Knocked down ErbB2 expression resulted in a similar inhibition of uPA and MMP-9 expression as noted by DHA and AG1478. Taken together, these results suggest that suppression of EGF-induced metastasis by DHA is likely through an inhibition of EGFR and ErbB2 protein expression and the downstream target uPA and MMP-9 activation in SK-BR3 human breast cancer cells.
Collapse
Affiliation(s)
- Chien-Chun Li
- a School of Nutrition, Chung Shan Medical University , Taichung , Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Goichberg P, Chang J, Liao R, Leri A. Cardiac stem cells: biology and clinical applications. Antioxid Redox Signal 2014; 21:2002-17. [PMID: 24597850 PMCID: PMC4208604 DOI: 10.1089/ars.2014.5875] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Heart disease is the primary cause of death in the industrialized world. Cardiac failure is dictated by an uncompensated reduction in the number of viable and fully functional cardiomyocytes. While current pharmacological therapies alleviate the symptoms associated with cardiac deterioration, heart transplantation remains the only therapy for advanced heart failure. Therefore, there is a pressing need for novel therapeutic modalities. Cell-based therapies involving cardiac stem cells (CSCs) constitute a promising emerging approach for the replenishment of the lost tissue and the restoration of cardiac contractility. RECENT ADVANCES CSCs reside in the adult heart and govern myocardial homeostasis and repair after injury by producing new cardiomyocytes and vascular structures. In the last decade, different classes of immature cells expressing distinct stem cell markers have been identified and characterized in terms of their growth properties, differentiation potential, and regenerative ability. Phase I clinical trials, employing autologous CSCs in patients with ischemic cardiomyopathy, are being completed with encouraging results. CRITICAL ISSUES Accumulating evidence concerning the role of CSCs in heart regeneration imposes a reconsideration of the mechanisms of cardiac aging and the etiology of heart failure. Deciphering the molecular pathways that prevent activation of CSCs in their environment and understanding the processes that affect CSC survival and regenerative function with cardiac pathologies, commonly accompanied by alterations in redox conditions, are of great clinical importance. FUTURE DIRECTIONS Further investigations of CSC biology may be translated into highly effective and novel therapeutic strategies aiming at the enhancement of the endogenous healing capacity of the diseased heart.
Collapse
Affiliation(s)
- Polina Goichberg
- Departments of Anesthesia and Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | | | | | | |
Collapse
|
28
|
Valente M, Nascimento DS, Cumano A, Pinto-do-Ó P. Sca-1+ cardiac progenitor cells and heart-making: a critical synopsis. Stem Cells Dev 2014; 23:2263-73. [PMID: 24926741 DOI: 10.1089/scd.2014.0197] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The identification, in the adult, of cardiomyocyte turnover events and of cardiac progenitor cells (CPCs) has revolutionized the field of cardiovascular medicine. However, the low rate of CPCs differentiation events reported both in vitro and in vivo, even after injury, raised concerns on the biological significance of these subsets. In this Comprehensive Review, we discuss the current understanding of cardiac Lin(-)Sca-1(+) cells in light of what is also known for cellular compartments with similar phenotypes in other organs. The Lin(-)Sca-1(+) heart subset is heterogeneous and displays a mesenchymal profile, characterized by a limited ability to generate cardiomyocytes in vitro and in vivo, even after injury. There is no evidence for Sca-1 expression in embryonic cardiovascular progenitors. In other organs, Sca-1 expression is mainly observed on mesoderm-derived cells, although it is not restricted to stem/progenitor cell populations. It is urgent to determine, at a single cell level, to which extent cardiac Lin(-)Sca-1(+) cells overlap with the fibroblast compartment.
Collapse
Affiliation(s)
- Mariana Valente
- 1 Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironments for NewTherapies Group, INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto , Porto, Portugal
| | | | | | | |
Collapse
|
29
|
Promotion of cardiac differentiation of brown adipose derived stem cells by chitosan hydrogel for repair after myocardial infarction. Biomaterials 2014; 35:3986-98. [PMID: 24508080 DOI: 10.1016/j.biomaterials.2014.01.021] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/08/2014] [Indexed: 02/03/2023]
Abstract
The ability to restore heart function by replacement of diseased myocardium is one of the great challenges in biomaterials and regenerative medicine. Brown adipose derived stem cells (BADSCs) present a new source of cardiomyocytes to regenerate the myocardium after infarction. In this study, we explored an injectable tissue engineering strategy to repair damaged myocardium, in which chitosan hydrogels were investigated as a carrier for BADSCs. In vitro, the effect and mechanism of chitosan components on the cardiac differentiation of BADSCs were investigated. In vivo, BADSCs carrying double-fusion reporter gene (firefly luciferase and monomeric red fluorescent protein (fluc-mRFP)) were transplanted into infarcted rat hearts with or without chitosan hydrogel. Multi-techniques were used to assess the effects of treatments. We observed that chitosan components significantly enhanced cardiac differentiation of BADSCs, which was assessed by percentages of cTnT(+) cells and expression of cardiac-specific markers, including GATA-4, Nkx2.5, Myl7, Myh6, cTnI, and Cacna1a. Treatment with collagen synthesis inhibitors, cis-4-hydroxy-D-proline (CIS), significantly inhibited the chitosan-enhanced cardiac differentiation, indicating that the enhanced collagen synthesis by chitosan accounts for its promotive role in cardiac differentiation of BADSCs. Longitudinal in vivo bioluminescence imaging and histological staining revealed that chitosan enhanced the survival of engrafted BADSCs and significantly increased the differentiation rate of BADSCs into cardiomyocytes in vivo. Furthermore, BADSCs delivered by chitosan hydrogel prevented adverse matrix remodeling, increased angiogenesis, and preserved heart function. These results suggested that the injectable cardiac tissue engineering based on chitosan hydrogel and BADSCs is a useful strategy for myocardium regeneration.
Collapse
|
30
|
Konstandin MH, Toko H, Gastelum GM, Quijada P, De La Torre A, Quintana M, Collins B, Din S, Avitabile D, Völkers M, Gude N, Fässler R, Sussman MA. Fibronectin is essential for reparative cardiac progenitor cell response after myocardial infarction. Circ Res 2013; 113:115-25. [PMID: 23652800 DOI: 10.1161/circresaha.113.301152] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Adoptive transfer of cardiac progenitor cells (CPCs) has entered clinical application, despite limited mechanistic understanding of the endogenous response after myocardial infarction (MI). Extracellular matrix undergoes dramatic changes after MI and therefore might be linked to CPC-mediated repair. OBJECTIVE To demonstrate the significance of fibronectin (Fn), a component of the extracellular matrix, for induction of the endogenous CPC response to MI. METHODS AND RESULTS This report shows that presence of CPCs correlates with the expression of Fn during cardiac development and after MI. In vivo, genetic conditional ablation of Fn blunts CPC response measured 7 days after MI through reduced proliferation and diminished survival. Attenuated vasculogenesis and cardiogenesis during recovery were evident at the end of a 12-week follow-up period. Impaired CPC-dependent reparative remodeling ultimately leads to continuous decline of cardiac function in Fn knockout animals. In vitro, Fn protects and induces proliferation of CPCs via β₁-integrin-focal adhesion kinase-signal transducer and activator of transcription 3-Pim1 independent of Akt. CONCLUSIONS Fn is essential for endogenous CPC expansion and repair required for stabilization of cardiac function after MI.
Collapse
Affiliation(s)
- Mathias H Konstandin
- San Diego State University Integrated Regenerative Research Institute, San Diego, CA 92182, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chung C, Pruitt BL, Heilshorn SC. Spontaneous cardiomyocyte differentiation of mouse embryoid bodies regulated by hydrogel crosslink density. Biomater Sci 2013; 1:1082-1090. [PMID: 24748962 DOI: 10.1039/c3bm60139k] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cellular therapies have great potential to provide alternative treatment options for those suffering from heart disease. In order to optimize cell delivery for therapeutic efficacy, a greater understanding of parameters that impact stem cell differentiation, survival, growth, and development are needed. In this study, we examine the role of hydrogel crosslink density on spontaneous cardiomyocyte (CM) differentiation of murine embryoid bodies (EBs). CM differentiation was accelerated in hydrogels of low crosslink density, where 100% of the hydrogels were positive for CM differentiation compared to only 53% in the high crosslink density group after 8 days of culture. DNA microarray data suggests that enhanced CM differentiation in the low crosslink density hydrogels was not tissue specific but rather a result of favoured EB development and cell proliferation. Additionally, enhanced EB growth and differentiation in low crosslink density hydrogels was independent of RGD ligand density and not a consequence of enhanced diffusion. We also demonstrate that matrix metalloproteinase activity is required for spontaneous CM differentiation in 3D hydrogels. Low hydrogel crosslink density regulates spontaneous EB differentiation by promoting EB growth and development. Elucidating the effects of microenvironmental cues on cell differentiation can aid in the optimization of stem cell-based therapies for tissue regeneration.
Collapse
Affiliation(s)
- Cindy Chung
- Materials Science and Engineering, McCullough Building, 476 Lomita Mall, Stanford, CA, USA. ; Tel: 650 723 3763 ; Mechanical Engineering, Durand Building, 496 Lomita Mall, Stanford, CA, USA
| | - Beth L Pruitt
- Mechanical Engineering, Durand Building, 496 Lomita Mall, Stanford, CA, USA
| | - Sarah C Heilshorn
- Materials Science and Engineering, McCullough Building, 476 Lomita Mall, Stanford, CA, USA. ; Tel: 650 723 3763
| |
Collapse
|
32
|
Drewa T, Joachimiak R, Bajek A, Gagat M, Grzanka A, Bodnar M, Marszalek A, Dębski R, Chłosta P. Hair follicle stem cells can be driven into a urothelial-like phenotype: an experimental study. Int J Urol 2012; 20:537-42. [PMID: 23088347 DOI: 10.1111/j.1442-2042.2012.03202.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 09/19/2012] [Indexed: 12/16/2022]
Abstract
The aim of this study was to show that conditioned medium might induce transdifferentiation of hair follicle stem cells into urothelial-like cells. Several conditioned media and culture conditions (skeletal muscle cell conditioned medium, smooth muscle cell conditioned medium, fibroblast conditioned medium, transforming growth factor-conditioned medium, urothelial cell conditioned medium, and co-culture of hair follicle stem cells and urothelial cells) were used. The hair follicle stem cells phenotype from rat whisker hair follicles was checked by using flow cytometry and immunofluorescence. Cytokeratins 7, 8, 15 and 18 were used as markers. Urothelial cell conditioned medium increased the expression of urothelial markers (cytokeratin 7, cytokeratin 8, cytokeratin 18), whereas it decreased a hair follicle stem cells marker (cytokeratin 15) after 2 weeks of culture. This process depended on the time of cultivation. This medium was able to sustain the epithelial phenotype of the culture. Other media including a co-culture system failed to induce similar changes. Smooth muscle conditioned medium resulted in a loss of cells in culture. Hair follicle stem cells are capable of differentiating into urothelial-like cells in vitro when exposed to a bladder-specific microenvironment.
Collapse
Affiliation(s)
- Tomasz Drewa
- Tissue Engineering Department, Nicolaus Copernicus University, Bydgoszcz, Poland.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bayomy AF, Bauer M, Qiu Y, Liao R. Regeneration in heart disease-Is ECM the key? Life Sci 2012; 91:823-7. [PMID: 22982346 DOI: 10.1016/j.lfs.2012.08.034] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/21/2012] [Accepted: 08/30/2012] [Indexed: 12/17/2022]
Abstract
The heart possesses a regeneration potential derived from endogenous and exogenous stem and progenitor cell populations, though baseline regeneration appears to be sub-therapeutic. This limitation was initially attributed to a lack of cells with cardiomyogenic potential following an insult to the myocardium. Rather, recent studies demonstrate increased numbers of cardiomyocyte progenitor cells in diseased hearts. Given that the limiting factor does not appear to be cell quantity but rather repletion of functional cardiomyocytes, it is crucial to understand potential mechanisms inhibiting progenitor cell differentiation. One of the extensively studied areas in heart disease is extracellular matrix (ECM) remodeling, with both the composition and mechanical properties of the ECM undergoing changes in diseased hearts. This review explores the influence of ECM properties on cardiomyogenesis and adult cardiac progenitor cells.
Collapse
Affiliation(s)
- Ahmad F Bayomy
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|