1
|
He H, Huang W, Zhang S, Li J, Zhang J, Li B, Xu J, Luo Y, Shi H, Li Y, Xiao J, Ezekiel OC, Li X, Wu J. Microneedle Patch for Transdermal Sequential Delivery of KGF-2 and aFGF to Enhance Burn Wound Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307485. [PMID: 38623988 DOI: 10.1002/smll.202307485] [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: 08/28/2023] [Revised: 12/19/2023] [Indexed: 04/17/2024]
Abstract
Severe burn wounds usually destroy key cells' functions of the skin resulting in delayed re-epithelization and wound regeneration. Promoting key cells' activities is crucial for burn wound repair. It is well known that keratinocyte growth factor-2 (KGF-2) participates in the proliferation and morphogenesis of epithelial cells while acidic fibroblast growth factor (aFGF) is a key mediator for fibroblast and endothelial cell growth and differentiation. However, thick eschar and the harsh environment of a burn wound often decrease the delivery efficiency of fibroblast growth factor (FGF) to the wound site. Therefore, herein a novel microneedle patch for sequential transdermal delivery of KGF-2 and aFGF is fabricated to enhance burn wound therapy. aFGF is first loaded in the nanoparticle (NPaFGF) and then encapsulated NPaFGF with KGF-2 in the microneedle patch (KGF-2/NPaFGF@MN). The result shows that KGF-2/NPaFGF@MN can successfully get across the eschar and sequentially release KGF-2 and aFGF. Additional data demonstrated that KGF-2/NPaFGF@MN achieved a quicker wound closure rate with reduced necrotic tissues, faster re-epithelialization, enhanced collagen deposition, and increased neo-vascularization. Further evidence suggests that improved wound healing is regulated by significantly elevated expressions of hypoxia-inducible factor-1 alpha (HIF-1ɑ) and heat shock protein 90 (Hsp90) in burn wounds. All these data proved that KGF-2/NPaFGF@MN is an effective treatment for wound healing of burns.
Collapse
Affiliation(s)
- Huacheng He
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, P. R. China
| | - Wen Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Shihui Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jie Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jian Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Bingxin Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jie Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Yuting Luo
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Huiling Shi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Yue Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Odinaka Cassandra Ezekiel
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jiang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| |
Collapse
|
2
|
Chen TA, Sharma D, Jia W, Ha D, Man K, Zhang J, Yang Y, Zhou Y, Kamp TJ, Zhao F. Detergent-Based Decellularization for Anisotropic Cardiac-Specific Extracellular Matrix Scaffold Generation. Biomimetics (Basel) 2023; 8:551. [PMID: 37999192 PMCID: PMC10669368 DOI: 10.3390/biomimetics8070551] [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: 08/09/2023] [Revised: 09/19/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Cell-derived extracellular matrix (ECM) has become increasingly popular in tissue engineering applications due to its ability to provide tailored signals for desirable cellular responses. Anisotropic cardiac-specific ECM scaffold decellularized from human induced pluripotent stem cell (hiPSC)-derived cardiac fibroblasts (hiPSC-CFs) mimics the native cardiac microenvironment and provides essential biochemical and signaling cues to hiPSC-derived cardiomyocytes (hiPSC-CMs). The objective of this study was to assess the efficacy of two detergent-based decellularization methods: (1) a combination of ethylenediaminetetraacetic acid and sodium dodecyl sulfate (EDTA + SDS) and (2) a combination of sodium deoxycholate and deoxyribonuclease (SD + DNase), in preserving the composition and bioactive substances within the aligned ECM scaffold while maximumly removing cellular components. The decellularization effects were evaluated by characterizing the ECM morphology, quantifying key structural biomacromolecules, and measuring preserved growth factors. Results showed that both treatments met the standard of cell removal (less than 50 ng/mg ECM dry weight) and substantially preserved major ECM biomacromolecules and growth factors. The EDTA + SDS treatment was more time-efficient and has been determined to be a more efficient method for generating an anisotropic ECM scaffold from aligned hiPSC-CFs. Moreover, this cardiac-specific ECM has demonstrated effectiveness in supporting the alignment of hiPSC-CMs and their expression of mature structural and functional proteins in in vitro cultures, which is crucial for cardiac tissue engineering.
Collapse
Affiliation(s)
- Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Wenkai Jia
- 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
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Jianhua Zhang
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Timothy J. Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
3
|
Rosellini E, Cascone MG, Guidi L, Schubert DW, Roether JA, Boccaccini AR. Mending a broken heart by biomimetic 3D printed natural biomaterial-based cardiac patches: a review. Front Bioeng Biotechnol 2023; 11:1254739. [PMID: 38047285 PMCID: PMC10690428 DOI: 10.3389/fbioe.2023.1254739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/16/2023] [Indexed: 12/05/2023] Open
Abstract
Myocardial infarction is one of the major causes of mortality as well as morbidity around the world. Currently available treatment options face a number of drawbacks, hence cardiac tissue engineering, which aims to bioengineer functional cardiac tissue, for application in tissue repair, patient specific drug screening and disease modeling, is being explored as a viable alternative. To achieve this, an appropriate combination of cells, biomimetic scaffolds mimicking the structure and function of the native tissue, and signals, is necessary. Among scaffold fabrication techniques, three-dimensional printing, which is an additive manufacturing technique that enables to translate computer-aided designs into 3D objects, has emerged as a promising technique to develop cardiac patches with a highly defined architecture. As a further step toward the replication of complex tissues, such as cardiac tissue, more recently 3D bioprinting has emerged as a cutting-edge technology to print not only biomaterials, but also multiple cell types simultaneously. In terms of bioinks, biomaterials isolated from natural sources are advantageous, as they can provide exceptional biocompatibility and bioactivity, thus promoting desired cell responses. An ideal biomimetic cardiac patch should incorporate additional functional properties, which can be achieved by means of appropriate functionalization strategies. These are essential to replicate the native tissue, such as the release of biochemical signals, immunomodulatory properties, conductivity, enhanced vascularization and shape memory effects. The aim of the review is to present an overview of the current state of the art regarding the development of biomimetic 3D printed natural biomaterial-based cardiac patches, describing the 3D printing fabrication methods, the natural-biomaterial based bioinks, the functionalization strategies, as well as the in vitro and in vivo applications.
Collapse
Affiliation(s)
| | | | - Lorenzo Guidi
- Department of Civil and Industrial Engineering, University of Pisa, Pisa, Italy
| | - Dirk W. Schubert
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University (FAU), Erlangen, Germany
- Bavarian Polymer Institute (BPI), Erlangen, Germany
| | - Judith A. Roether
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University (FAU), Erlangen, Germany
| | - Aldo R. Boccaccini
- Bavarian Polymer Institute (BPI), Erlangen, Germany
- Department of Materials Science and Engineering, Institute of Biomaterials, Friedrich-Alexander-University (FAU), Erlangen, Germany
| |
Collapse
|
4
|
Urciuolo F, Imparato G, Netti PA. In vitro strategies for mimicking dynamic cell-ECM reciprocity in 3D culture models. Front Bioeng Biotechnol 2023; 11:1197075. [PMID: 37434756 PMCID: PMC10330728 DOI: 10.3389/fbioe.2023.1197075] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
The extracellular microenvironment regulates cell decisions through the accurate presentation at the cell surface of a complex array of biochemical and biophysical signals that are mediated by the structure and composition of the extracellular matrix (ECM). On the one hand, the cells actively remodel the ECM, which on the other hand affects cell functions. This cell-ECM dynamic reciprocity is central in regulating and controlling morphogenetic and histogenetic processes. Misregulation within the extracellular space can cause aberrant bidirectional interactions between cells and ECM, resulting in dysfunctional tissues and pathological states. Therefore, tissue engineering approaches, aiming at reproducing organs and tissues in vitro, should realistically recapitulate the native cell-microenvironment crosstalk that is central for the correct functionality of tissue-engineered constructs. In this review, we will describe the most updated bioengineering approaches to recapitulate the native cell microenvironment and reproduce functional tissues and organs in vitro. We have highlighted the limitations of the use of exogenous scaffolds in recapitulating the regulatory/instructive and signal repository role of the native cell microenvironment. By contrast, strategies to reproduce human tissues and organs by inducing cells to synthetize their own ECM acting as a provisional scaffold to control and guide further tissue development and maturation hold the potential to allow the engineering of fully functional histologically competent three-dimensional (3D) tissues.
Collapse
Affiliation(s)
- F. Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - G. Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - P. A. Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|
5
|
Magaud C, Harnois T, Sebille S, Chatelier A, Faivre JF, Bois P, Page G, Gellen B. Pro-inflammatory cytokine secretion induced by amyloid transthyretin in human cardiac fibroblasts. Biochem Biophys Res Commun 2023; 642:83-89. [PMID: 36566566 DOI: 10.1016/j.bbrc.2022.12.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Extracellular aggregates of wild-type human transthyretin are associated with heart diseases such as wild-type transthyretin (TTR)-derived amyloidosis (ATTR-wt). Due to their strategic location, cardiac fibroblasts act as sentinel cells that sense injury and activate the inflammasome. No studies of the effects of TTR amyloid aggregation on the secretion of inflammatory factors by primary human cardiac fibroblasts (hCFs) have been reported yet. The intracellular internalization of TTR aggregates, which correspond to the early stage of ATTR-wt, were determined using immunofluorescence and Western blotting of cell lysates. A further objective of this study was to analyze the secretion of inflammatory factors by hCFs after analysis of TTR amyloid aggregation using X-MAP® Luminex Assay techniques. We show that TTR aggregates are internalized in hCFs and induce the secretion of both Brain Natriuretic Peptide (BNP) and N-terminal pro B-type Natriuretic Peptide(NT-proBNP). Also, pro-inflammatory mediators such as interleukin-6 (IL-6) and IL-8 are secreted without significant changes in the levels of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs). In conclusion, these findings suggest that IL-6 and IL-8 play important roles in the development of ATTR-wt, and indicate that IL-6 in particular could be a potentially important therapeutic target in patients with ATTR-wt.
Collapse
Affiliation(s)
| | - Thomas Harnois
- Laboratoire 4CS UMR 6041 CNRS, Université de Poitiers, France
| | | | | | | | - Patrick Bois
- Laboratoire PRéTI UR 24184, Université de Poitiers, France.
| | - Guylene Page
- UFR Médecine et Pharmacie, Université de Poitiers, France
| | | |
Collapse
|
6
|
Parboiled Germinated Brown Rice Improves Cardiac Structure and Gene Expression in Hypertensive Rats. Foods 2022; 12:foods12010009. [PMID: 36613225 PMCID: PMC9818593 DOI: 10.3390/foods12010009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/30/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hypertension leads to oxidative stress, inflammation, and fibrosis. The suppression of these indicators may be one treatment approach. Parboiled germinated brown rice (PGBR), obtained by steaming germinated Jasmine rice, reduces oxidative stress and inflammation in vivo. PGBR contains more bioactive compounds than brown rice (BR) and white rice (WR). Anti-hypertensive benefits of PGBR have been predicted, but research is lacking. The anti-hypertensive effects of PGBR were investigated in the downstream gene network of hypertension pathogenesis, including the renin-angiotensin system, fibrosis, oxidative stress production, and antioxidant enzymes in N-nitro-L-arginine methyl ester (L-NAME)-induced hypertensive rats. To strengthen our findings, the cardiac structure was also studied. PGBR-exposed rats showed significant reductions in systolic blood pressure (SBP) compared to the hypertensive group. WR did not reduce SBP because of the loss of bioactive compounds during intensive milling. PGBR also reduced the expression of the angiotensin type 1 receptor (AT1R), transforming growth factor-β (TGF-β), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX4), which contribute to the renin-angiotensin system, fibrosis, and oxidative stress production, respectively. Losartan (Los, an anti-hypertensive drug)-treated rats also exhibited similar gene expression, implying that PGBR may reduce hypertension using the same downstream target as Los. Our data also indicated that PGBR reduced cardiac lesions, such as the cardiomyopathy induced by L-NAME. This is the first report on the anti-hypertensive effects of PGBR in vivo by the suppression of the renin response, fibrosis, and improved cardiac structure.
Collapse
|
7
|
Dickerson DA. Advancing Engineered Heart Muscle Tissue Complexity with Hydrogel Composites. Adv Biol (Weinh) 2022; 7:e2200067. [PMID: 35999488 DOI: 10.1002/adbi.202200067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/19/2022] [Indexed: 11/10/2022]
Abstract
A heart attack results in the permanent loss of heart muscle and can lead to heart disease, which kills more than 7 million people worldwide each year. To date, outside of heart transplantation, current clinical treatments cannot regenerate lost heart muscle or restore full function to the damaged heart. There is a critical need to create engineered heart tissues with structural complexity and functional capacity needed to replace damaged heart muscle. The inextricable link between structure and function suggests that hydrogel composites hold tremendous promise as a biomaterial-guided strategy to advance heart muscle tissue engineering. Such composites provide biophysical cues and functionality as a provisional extracellular matrix that hydrogels cannot on their own. This review describes the latest advances in the characterization of these biomaterial systems and using them for heart muscle tissue engineering. The review integrates results across the field to provide new insights on critical features within hydrogel composites and perspectives on the next steps to harnessing these promising biomaterials to faithfully reproduce the complex structure and function of native heart muscle.
Collapse
Affiliation(s)
- Darryl A. Dickerson
- Department of Mechanical and Materials Engineering Florida International University 10555 West Flagler St Miami FL 33174 USA
| |
Collapse
|
8
|
Zivari-Ghader T, Dolati S, Mehdizadeh A, Davaran S, Rashidi MR, Yousefi M. Recent scaffold-based tissue engineering approaches in premature ovarian failure treatment. J Tissue Eng Regen Med 2022; 16:605-620. [PMID: 35511799 DOI: 10.1002/term.3306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022]
Abstract
Recently, tissue engineering and regenerative medicine have received significant attention with outstanding advances. The main scope of this technology is to recover the damaged tissues and organs or to maintain and improve their function. One of the essential fields in tissue engineering is scaffold designing and construction, playing an integral role in damaged tissues reconstruction and repair. However, premature ovarian failure (POF) is a disorder causing many medical and psychological problems in women. POF treatment using tissue engineering and various scaffold has recently made tremendous and promising progress. Due to the importance of the subject, we have summarized the recently examined scaffolds in the treatment of POF in this review.
Collapse
Affiliation(s)
- Tayyebeh Zivari-Ghader
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Ambade AS, Hassoun PM, Damico RL. Basement Membrane Extracellular Matrix Proteins in Pulmonary Vascular and Right Ventricular Remodeling in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2021; 65:245-258. [PMID: 34129804 PMCID: PMC8485997 DOI: 10.1165/rcmb.2021-0091tr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM), a highly organized network of structural and nonstructural proteins, plays a pivotal role in cellular and tissue homeostasis. Changes in the ECM are critical for normal tissue repair, whereas dysregulation contributes to aberrant tissue remodeling. Pulmonary arterial hypertension is a severe disorder of the pulmonary vasculature characterized by pathologic remodeling of the pulmonary vasculature and right ventricle, increased production and deposition of structural and nonstructural proteins, and altered expression of ECM growth factors and proteases. Furthermore, ECM remodeling plays a significant role in disease progression, as several dynamic changes in its composition, quantity, and organization are documented in both humans and animal models of disease. These ECM changes impact vascular cell biology and affect proliferation of resident cells. Furthermore, ECM components determine the tissue architecture of the pulmonary and myocardial vasculature as well as the myocardium itself and provide mechanical stability crucial for tissue homeostasis. However, little is known about the basement membrane (BM), a specialized, self-assembled conglomerate of ECM proteins, during remodeling. In the vasculature, the BM is in close physical association with the vascular endothelium and smooth muscle cells. While in the myocardium, each cardiomyocyte is enclosed by a BM that serves as the interface between cardiomyocytes and the surrounding interstitial matrix. In this review, we provide a brief overview on the current state of knowledge of the BM and its ECM composition and their impact on pulmonary vascular remodeling and right ventricle dysfunction and failure in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anjira S Ambade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
10
|
Mannino G, Russo C, Maugeri G, Musumeci G, Vicario N, Tibullo D, Giuffrida R, Parenti R, Lo Furno D. Adult stem cell niches for tissue homeostasis. J Cell Physiol 2021; 237:239-257. [PMID: 34435361 PMCID: PMC9291197 DOI: 10.1002/jcp.30562] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
Adult stem cells are fundamental to maintain tissue homeostasis, growth, and regeneration. They reside in specialized environments called niches. Following activating signals, they proliferate and differentiate into functional cells that are able to preserve tissue physiology, either to guarantee normal turnover or to counteract tissue damage caused by injury or disease. Multiple interactions occur within the niche between stem cell‐intrinsic factors, supporting cells, the extracellular matrix, and signaling pathways. Altogether, these interactions govern cell fate, preserving the stem cell pool, and regulating stem cell proliferation and differentiation. Based on their response to body needs, tissues can be largely classified into three main categories: tissues that even in normal conditions are characterized by an impressive turnover to replace rapidly exhausting cells (blood, epidermis, or intestinal epithelium); tissues that normally require only a basal cell replacement, though able to efficiently respond to increased tissue needs, injury, or disease (skeletal muscle); tissues that are equipped with less powerful stem cell niches, whose repairing ability is not able to overcome severe damage (heart or nervous tissue). The purpose of this review is to describe the main characteristics of stem cell niches in these different tissues, highlighting the various components influencing stem cell activity. Although much has been done, more work is needed to further increase our knowledge of niche interactions. This would be important not only to shed light on this fundamental chapter of human physiology but also to help the development of cell‐based strategies for clinical therapeutic applications, especially when other approaches fail.
Collapse
Affiliation(s)
- Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
11
|
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: 105] [Impact Index Per Article: 26.3] [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
|
12
|
Mohammadi Nasr S, Rabiee N, Hajebi S, Ahmadi S, Fatahi Y, Hosseini M, Bagherzadeh M, Ghadiri AM, Rabiee M, Jajarmi V, Webster TJ. Biodegradable Nanopolymers in Cardiac Tissue Engineering: From Concept Towards Nanomedicine. Int J Nanomedicine 2020; 15:4205-4224. [PMID: 32606673 PMCID: PMC7314574 DOI: 10.2147/ijn.s245936] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/02/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the number one cause of heart failure and death in the world, and the transplantation of the heart is an effective and viable choice for treatment despite presenting many disadvantages (most notably, transplant heart availability). To overcome this problem, cardiac tissue engineering is considered a promising approach by using implantable artificial blood vessels, injectable gels, and cardiac patches (to name a few) made from biodegradable polymers. Biodegradable polymers are classified into two main categories: natural and synthetic polymers. Natural biodegradable polymers have some distinct advantages such as biodegradability, abundant availability, and renewability but have some significant drawbacks such as rapid degradation, insufficient electrical conductivity, immunological reaction, and poor mechanical properties for cardiac tissue engineering. Synthetic biodegradable polymers have some advantages such as strong mechanical properties, controlled structure, great processing flexibility, and usually no immunological concerns; however, they have some drawbacks such as a lack of cell attachment and possible low biocompatibility. Some applications have combined the best of both and exciting new natural/synthetic composites have been utilized. Recently, the use of nanostructured polymers and polymer nanocomposites has revolutionized the field of cardiac tissue engineering due to their enhanced mechanical, electrical, and surface properties promoting tissue growth. In this review, recent research on the use of biodegradable natural/synthetic nanocomposite polymers in cardiac tissue engineering is presented with forward looking thoughts provided for what is needed for the field to mature.
Collapse
Affiliation(s)
| | - Navid Rabiee
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Sakineh Hajebi
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, Iran
- Institute of Polymeric Materials, Sahand University of Technology, Tabriz, Iran
| | - Sepideh Ahmadi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Masoumehossadat Hosseini
- Department of Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, Tehran, Iran
- Soroush Mana Pharmed, Pharmaceutical Holding, Golrang Industrial Group, Tehran, Iran
| | | | | | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Vahid Jajarmi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA02115, United States
| |
Collapse
|
13
|
Abstract
Cardiac fibroblasts and fibrosis contribute to the pathogenesis of heart failure, a prevalent cause of mortality. Therefore, a majority of the existing information regarding cardiac fibroblasts is focused on their function and behavior after heart injury. Less is understood about the signaling and transcriptional networks required for the development and homeostatic roles of these cells. This review is devoted to describing our current understanding of cardiac fibroblast development. I detail cardiac fibroblast formation during embryogenesis including the discovery of a second embryonic origin for cardiac fibroblasts. Additional information is provided regarding the roles of the genes essential for cardiac fibroblast development. It should be noted that many questions remain regarding the cell-fate specification of these fibroblast progenitors, and it is hoped that this review will provide a basis for future studies regarding this topic.
Collapse
|
14
|
Alassaf A, Tansik G, Mayo V, Wubker L, Carbonero D, Agarwal A. Engineering anisotropic cardiac monolayers on microelectrode arrays for non-invasive analyses of electrophysiological properties. Analyst 2020; 145:139-149. [DOI: 10.1039/c9an01339c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Engineering cardiac tissues with physiological architectural and mechanical properties on microelectrode arrays enables long term culture and non-invasive collection of electrophysiological readouts.
Collapse
Affiliation(s)
- Ahmad Alassaf
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Department of Medical Equipment Technology
| | - Gulistan Tansik
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Vera Mayo
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Laura Wubker
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Daniel Carbonero
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering
- University of Miami
- Coral Gables
- USA
- Dr. John T Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami
| |
Collapse
|
15
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
16
|
Burke MJ, Walmsley R, Munsey TS, Smith AJ. Receptor tyrosine kinase inhibitors cause dysfunction in adult rat cardiac fibroblasts in vitro. Toxicol In Vitro 2019; 58:178-186. [DOI: 10.1016/j.tiv.2019.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/28/2022]
|
17
|
Sykora M, Kamocsaiova L, Egan Benova T, Frimmel K, Ujhazy E, Mach M, Barancik M, Tribulova N, Szeiffova Bacova B. Alterations in myocardial connexin-43 and matrix metalloproteinase-2 signaling in response to pregnancy and oxygen deprivation of Wistar rats: a pilot study 1. Can J Physiol Pharmacol 2019; 97:829-836. [PMID: 30908945 DOI: 10.1139/cjpp-2018-0740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two important aspects of cardiac adaptive response to pregnancy have been studied in normal as well as hypoxic conditions: (1) intercellular signaling mediated by myocardial connexin-43 (Cx43) that is crucial to synchronize heart function; (2) extracellular signaling mediated by matrix metalloproteinase-2 (MMP-2) that is an early marker of extracellular matrix remodeling. Myocardial Cx43 distribution and functional capillary density were determined as well. Hypoxia was induced by exposure of rats to 10.5% O2 and 89.5% N2 in a hermetically sealed chamber. Findings showed that pregnancy resulted in a significant increase of Cx43 protein expression, its functional phosphorylated forms, and enhanced capillary density while did not affect either expression of total MMP-2 or its activity. Maternal hypoxia for 12 or 16 h did not affect elevated Cx43 but enhanced its distribution on lateral sides of the cardiomyocytes. In contrast, hypoxia of nonpregnant rats resulted in upregulation of Cx43, its lateral distribution, and enhanced capillary density. Hypoxia did not affect myocardial MMP-2 either in pregnant or nonpregnant rats. Cardiac adaptive response to pregnancy is accompanied by enhanced Cx43 without changes in MMP-2 signaling. Pregnant rat heart is tolerant to short-term hypoxemia, while nonpregnant rat heart reacts by upregulation of Cx43 and increased capillary density.
Collapse
Affiliation(s)
- Matus Sykora
- CEM SAS, Institute for Heart Research, Bratislava, Slovakia
| | - Lucia Kamocsaiova
- Faculty of Natural Sciences of Comenius University, Bratislava, Slovakia
| | | | - Karel Frimmel
- CEM SAS, Institute for Heart Research, Bratislava, Slovakia
| | - Eduard Ujhazy
- CEM SAS, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovakia
| | - Mojmir Mach
- CEM SAS, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovakia
| | | | | | | |
Collapse
|
18
|
Novaes RD, Mouro VGS, Gonçalves RV, Mendonça AAS, Santos EC, Fialho MCQ, Machado-Neves M. Aluminum: A potentially toxic metal with dose-dependent effects on cardiac bioaccumulation, mineral distribution, DNA oxidation and microstructural remodeling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:814-826. [PMID: 30032078 DOI: 10.1016/j.envpol.2018.07.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/23/2018] [Accepted: 07/09/2018] [Indexed: 06/08/2023]
Abstract
Large amounts of aluminum (Al) are found in wastewater from industrial bauxite mining, which is often responsible for the contamination of drinking water sources in urban and rural communities. Although this metal exhibits broad environmental distribution, its cardiac repercussions are poorly understood, making it difficult to establish diagnostic criteria in cases of Al intoxication. In the absence of clinical data, we used a preclinical model to investigate the impact of Al exposure on heart bioaccumulation, molecular oxidation, micromineral distribution, structural and ultrastructural remodeling of the cardiac tissue. Male Wistar rats were equally randomized into five groups: G1 = distilled water; and G2 to G5 = 0.02, 0.1, 50, and 200 mg/kg aluminum solution, respectively. After 120 days, the hearts were collected and subjected to mineral microanalysis, immunoenzymatic detection of 8-OHdG, as well as bright field, polarizing, scanning and transmission electron microscopy to estimate the extent of the cardiac remodeling and cardiomyocytes ultrastructure. Long-term Al exposure induced dose-dependent bioaccumulation, micromineral imbalance, genomic DNA oxidation, structural and ultrastructural abnormalities of the cardiac tissue, resulting in extensive parenchymal loss, stromal expansion, diffuse inflammatory infiltrate, increased glycoconjugate and collagen deposition, subversion and collapse of the collagen network, reduced myocardial vascularization index, mitochondrial swelling, sarcomere disorganization, myofilament dissociation, and fragmentation in cardiomyocytes. Our findings indicated that the heart was sensitive to Al-mediated toxicity, especially in animals treated with the three highest doses of Al. In response to Al-induced loss of the parenchyma, heart stroma exhibited a reactive and compensatory expansion, which, in combination with the increased distribution of thick myofibrils and degenerated mitochondria in cardiomyocytes, provides morphological evidence that cardiac tissue adaptations are not enough to adjust the relationships between the parenchyma and stroma until a steady state is reached, resulting in continuous pathological remodeling potentially associated with Al-induced proinflammatory and pro-oxidant events.
Collapse
Affiliation(s)
- Rômulo D Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, MG, Brazil.
| | - Viviane G S Mouro
- Department of General Biology, Federal University of Viçosa, MG, Brazil
| | | | - Andrea A S Mendonça
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, MG, Brazil
| | - Eliziária C Santos
- Medicine School, Federal University of Jequitinhonha and Mucuri Valleys, MG, Brazil
| | - Maria C Q Fialho
- Department of Morphology, Federal University of Amazonas, AM, Brazil
| | | |
Collapse
|
19
|
Rosellini E, Zhang YS, Migliori B, Barbani N, Lazzeri L, Shin SR, Dokmeci MR, Cascone MG. Protein/polysaccharide-based scaffolds mimicking native extracellular matrix for cardiac tissue engineering applications. J Biomed Mater Res A 2018; 106:769-781. [PMID: 29052369 PMCID: PMC5845858 DOI: 10.1002/jbm.a.36272] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 11/07/2022]
Abstract
Tissue engineering has emerged as a viable approach to treat disease or repair damage in tissues and organs. One of the key elements for the success of tissue engineering is the use of a scaffold serving as artificial extracellular matrix (ECM). The ECM hosts the cells and improves their survival, proliferation, and differentiation, enabling the formation of new tissue. Here, we propose the development of a class of protein/polysaccharide-based porous scaffolds for use as ECM substitutes in cardiac tissue engineering. Scaffolds based on blends of a protein component, collagen or gelatin, with a polysaccharide component, alginate, were produced by freeze-drying and subsequent ionic and chemical crosslinking. Their morphological, physicochemical, and mechanical properties were determined and compared with those of natural porcine myocardium. We demonstrated that our scaffolds possessed highly porous and interconnected structures, and the chemical homogeneity of the natural ECM was well reproduced in both types of scaffolds. Furthermore, the alginate/gelatin (AG) scaffolds better mimicked the native tissue in terms of interactions between components and protein secondary structure, and in terms of swelling behavior. The AG scaffolds also showed superior mechanical properties for the desired application and supported better adhesion, growth, and differentiation of myoblasts under static conditions. The AG scaffolds were subsequently used for culturing neonatal rat cardiomyocytes, where high viability of the resulting cardiac constructs was observed under dynamic flow culture in a microfluidic bioreactor. We therefore propose our protein/polysaccharide scaffolds as a viable ECM substitute for applications in cardiac tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 769-781, 2018.
Collapse
Affiliation(s)
- Elisabetta Rosellini
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Bianca Migliori
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Niccoletta Barbani
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Luigi Lazzeri
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Mehmet Remzi Dokmeci
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Maria Grazia Cascone
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| |
Collapse
|
20
|
Besser RR, Ishahak M, Mayo V, Carbonero D, Claure I, Agarwal A. Engineered Microenvironments for Maturation of Stem Cell Derived Cardiac Myocytes. Am J Cancer Res 2018; 8:124-140. [PMID: 29290797 PMCID: PMC5743464 DOI: 10.7150/thno.19441] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Through the use of stem cell-derived cardiac myocytes, tissue-engineered human myocardial constructs are poised for modeling normal and diseased physiology of the heart, as well as discovery of novel drugs and therapeutic targets in a human relevant manner. This review highlights the recent bioengineering efforts to recapitulate microenvironmental cues to further the maturation state of newly differentiated cardiac myocytes. These techniques include long-term culture, co-culture, exposure to mechanical stimuli, 3D culture, cell-matrix interactions, and electrical stimulation. Each of these methods has produced various degrees of maturation; however, a standardized measure for cardiomyocyte maturation is not yet widely accepted by the scientific community.
Collapse
|
21
|
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
|
22
|
Effect of Botulinum Toxin Type A on TGF-β/Smad Pathway Signaling: Implications for Silicone-Induced Capsule Formation. Plast Reconstr Surg 2017; 138:821e-829e. [PMID: 27391832 DOI: 10.1097/prs.0000000000002625] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND One of the most serious complications of breast surgery using implants is capsular contracture. Several preventive treatments have been introduced; however, the mechanism of capsule formation has not been resolved completely. The authors previously identified negative effects of botulinum toxin type A on capsule formation, expression of transforming growth factor (TGF)-β1, and differentiation of fibroblasts into myofibroblasts. Thus, the authors investigated how to prevent capsule formation by using botulinum toxin type A, particularly by means of TGF-β1 signaling, in human fibroblasts. METHODS In vitro, cultured human fibroblasts were treated with TGF-β1 and/or botulinum toxin type A. Expression of collagen, matrix metalloproteinase, and Smad was examined by Western blotting. The activation of matrix metalloproteinase was observed by gelatin zymography. In vivo, the effect of botulinum toxin type A on the phosphorylation of Smad2 in silicone-induced capsule formation was evaluated by immunocytochemistry. RESULTS In vitro, the phosphorylation of Smad2 was inhibited by botulinum toxin type A treatment. The expression levels of collagen types 1 and 3 were inhibited by botulinum toxin type A treatment, whereas those of matrix metalloproteinase-2 and matrix metalloproteinase-9 were enhanced. Gelatin zymography experiments confirmed enhanced matrix metalloproteinase-2 activity in collagen degradation. In vivo, botulinum toxin type A treatment reduced capsule thickness and Smad2 phosphorylation in silicone-induced capsules. CONCLUSION This study suggests that botulinum toxin type A plays an important role in the inhibition of capsule formation through the TGF-β/Smad signaling pathway. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
23
|
Scuderi GJ, Butcher J. Naturally Engineered Maturation of Cardiomyocytes. Front Cell Dev Biol 2017; 5:50. [PMID: 28529939 PMCID: PMC5418234 DOI: 10.3389/fcell.2017.00050] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022] Open
Abstract
Ischemic heart disease remains one of the most prominent causes of mortalities worldwide with heart transplantation being the gold-standard treatment option. However, due to the major limitations associated with heart transplants, such as an inadequate supply and heart rejection, there remains a significant clinical need for a viable cardiac regenerative therapy to restore native myocardial function. Over the course of the previous several decades, researchers have made prominent advances in the field of cardiac regeneration with the creation of in vitro human pluripotent stem cell-derived cardiomyocyte tissue engineered constructs. However, these engineered constructs exhibit a functionally immature, disorganized, fetal-like phenotype that is not equivalent physiologically to native adult cardiac tissue. Due to this major limitation, many recent studies have investigated approaches to improve pluripotent stem cell-derived cardiomyocyte maturation to close this large functionality gap between engineered and native cardiac tissue. This review integrates the natural developmental mechanisms of cardiomyocyte structural and functional maturation. The variety of ways researchers have attempted to improve cardiomyocyte maturation in vitro by mimicking natural development, known as natural engineering, is readily discussed. The main focus of this review involves the synergistic role of electrical and mechanical stimulation, extracellular matrix interactions, and non-cardiomyocyte interactions in facilitating cardiomyocyte maturation. Overall, even with these current natural engineering approaches, pluripotent stem cell-derived cardiomyocytes within three-dimensional engineered heart tissue still remain mostly within the early to late fetal stages of cardiomyocyte maturity. Therefore, although the end goal is to achieve adult phenotypic maturity, more emphasis must be placed on elucidating how the in vivo fetal microenvironment drives cardiomyocyte maturation. This information can then be utilized to develop natural engineering approaches that can emulate this fetal microenvironment and thus make prominent progress in pluripotent stem cell-derived maturity toward a more clinically relevant model for cardiac regeneration.
Collapse
Affiliation(s)
- Gaetano J Scuderi
- Meinig School of Biomedical Engineering, Cornell UniversityIthaca, NY, USA
| | - Jonathan Butcher
- Meinig School of Biomedical Engineering, Cornell UniversityIthaca, NY, USA
| |
Collapse
|
24
|
Targeted inhibition of Focal Adhesion Kinase Attenuates Cardiac Fibrosis and Preserves Heart Function in Adverse Cardiac Remodeling. Sci Rep 2017; 7:43146. [PMID: 28225063 PMCID: PMC5320468 DOI: 10.1038/srep43146] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/16/2016] [Indexed: 12/02/2022] Open
Abstract
Cardiac fibrosis in post-myocardial infarction (MI), seen in both infarcted and non-infarcted myocardium, is beneficial to the recovery of heart function. But progressively pathological fibrosis impairs ventricular function and leads to poor prognosis. FAK has recently received attention as a potential mediator of fibrosis, our previous study reported that pharmacological inhibition of FAK can attenuate cardiac fibrosis in post MI models. However, the long-term effects on cardiac function and adverse cardiac remodelling were not clearly investigated. In this study, we tried to determine the preliminary mechanisms in regulating CF transformation to myofibroblasts and ECM synthesis relevant to the development of adverse cardiac remolding in vivo and in vitro. Our study provides even more evidence that FAK is directly related to the activation of CF in hypoxia condition in a dose-dependent and time-dependent manner. Pharmacological inhibition of FAK significantly reduces myofibroblast differentiation; our in vivo data demonstrated that a FAK inhibitor significantly decreases fibrotic score, and preserves partial left ventricular function. Both PI3K/AKT signalling and ERK1/2 are necessary for hypoxia-induced CF differentiation and ECM synthesis; this process also involves lysyl oxidase (LOX). These findings suggest that pharmacological inhibition of FAK may become an effective therapeutic strategy against adverse fibrosis.
Collapse
|
25
|
Wanjare M, Huang NF. Regulation of the microenvironment for cardiac tissue engineering. Regen Med 2017; 12:187-201. [PMID: 28244821 DOI: 10.2217/rme-2016-0132] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The microenvironment of myocardium plays an important role in the fate and function of cardiomyocytes (CMs). Cardiovascular tissue engineering strategies commonly utilize stem cell sources in conjunction with microenvironmental cues that often include biochemical, electrical, spatial and biomechanical factors. Microenvironmental stimulation of CMs, in addition to the incorporation of intercellular interactions from non-CMs, results in the generation of engineered cardiac constructs. Current studies suggest that use of these factors when engineering cardiac constructs improve cardiac function when implanted in vivo. In this review, we summarize the approaches to modulate biochemical, electrical, biomechanical and spatial factors to induce CM differentiation and their subsequent organization for cardiac tissue engineering application.
Collapse
Affiliation(s)
- Maureen Wanjare
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ngan F Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
26
|
Deddens JC, Sadeghi AH, Hjortnaes J, van Laake LW, Buijsrogge M, Doevendans PA, Khademhosseini A, Sluijter JPG. Modeling the Human Scarred Heart In Vitro: Toward New Tissue Engineered Models. Adv Healthc Mater 2017; 6. [PMID: 27906521 DOI: 10.1002/adhm.201600571] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Cardiac remodeling is critical for effective tissue healing, however, excessive production and deposition of extracellular matrix components contribute to scarring and failing of the heart. Despite the fact that novel therapies have emerged, there are still no lifelong solutions for this problem. An urgent need exists to improve the understanding of adverse cardiac remodeling in order to develop new therapeutic interventions that will prevent, reverse, or regenerate the fibrotic changes in the failing heart. With recent advances in both disease biology and cardiac tissue engineering, the translation of fundamental laboratory research toward the treatment of chronic heart failure patients becomes a more realistic option. Here, the current understanding of cardiac fibrosis and the great potential of tissue engineering are presented. Approaches using hydrogel-based tissue engineered heart constructs are discussed to contemplate key challenges for modeling tissue engineered cardiac fibrosis and to provide a future outlook for preclinical and clinical applications.
Collapse
Affiliation(s)
- Janine C. Deddens
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
| | - Amir Hossein Sadeghi
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Biomaterials Innovation Research Center; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Sciences & Technology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Linda W. van Laake
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Marc Buijsrogge
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Sciences & Technology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Wyss Institute for Biologically Inspired Engineering; Harvard University; Boston MA 02115 USA
- Department of Physics; King Abdulaziz University; Jeddah 21569 Saudi Arabia
| | - Joost P. G. Sluijter
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| |
Collapse
|
27
|
Liu WY, Lin SG, Zhuo RY, Xie YY, Pan W, Lin XF, Shen FX. Xenogeneic Decellularized Scaffold: A Novel Platform for Ovary Regeneration. Tissue Eng Part C Methods 2017; 23:61-71. [PMID: 27981878 DOI: 10.1089/ten.tec.2016.0410] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Women younger than 40 years may face early menopause because of premature ovarian failure (POF). The cause of POF can be idiopathic or iatrogenic, especially the cancer-induced oophorectomy and chemo- or radiation therapy. The current treatments, including hormone replacement therapy (HRT) and cryopreservation techniques, have increased risk of ovarian cancer and may reintroduce malignant cells after autografting. Decellularization technique has been regarded as a novel regenerative medicine strategy for organ replacement, wherein the living cells of an organ are removed, leaving the extracellular matrix (ECM) for cellular seeding. This study aimed to produce a xenogeneic decellularized ovary (D-ovary) scaffold as a platform for ovary regeneration and transplantation. We have developed a novel decellularization protocol for porcine ovary by treatment with physical, chemical, and enzymatic methods. Using hematoxylin and eosin (H&E) staining, DAPI staining, scanning electron microscopy (SEM), and quantitative analysis, this approach proved effective in removing cellular components and preserving ECM. Furthermore, the results of biological safety evaluation demonstrated that the D-ovary tissues were noncytotoxic for rat ovarian cells in vitro and caused only a minimal immunogenic response in vivo. In addition, the D-ovary tissues successfully supported rat granulosa cell penetration ex vivo and showed an improvement in estradiol (E2) hormone secretion.
Collapse
Affiliation(s)
- Wen-Yue Liu
- 1 Department of Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
| | - Shi-Gang Lin
- 2 School of the First Clinical Medical Sciences, Wenzhou Medical University , Wenzhou, China
| | - Ru-Yi Zhuo
- 1 Department of Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
| | - Yuan-Yuan Xie
- 1 Department of Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
| | - Wei Pan
- 1 Department of Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
| | - Xian-Feng Lin
- 3 Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University , Hangzhou, China
| | - Fei-Xia Shen
- 1 Department of Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, China
| |
Collapse
|
28
|
Chacar S, Farès N, Bois P, Faivre JF. Basic Signaling in Cardiac Fibroblasts. J Cell Physiol 2016; 232:725-730. [DOI: 10.1002/jcp.25624] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Stéphanie Chacar
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Nassim Farès
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| | - Jean-François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| |
Collapse
|
29
|
Aminkeng F, Ross CJD, Rassekh SR, Hwang S, Rieder MJ, Bhavsar AP, Smith A, Sanatani S, Gelmon KA, Bernstein D, Hayden MR, Amstutz U, Carleton BC. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol 2016; 82:683-95. [PMID: 27197003 PMCID: PMC5338111 DOI: 10.1111/bcp.13008] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022] Open
Abstract
AIMS Anthracycline-induced cardiotoxicity (ACT) occurs in 57% of treated patients and remains an important limitation of anthracycline-based chemotherapy. In various genetic association studies, potential genetic risk markers for ACT have been identified. Therefore, we developed evidence-based clinical practice recommendations for pharmacogenomic testing to further individualize therapy based on ACT risk. METHODS We followed a standard guideline development process, including a systematic literature search, evidence synthesis and critical appraisal, and the development of clinical practice recommendations with an international expert group. RESULTS RARG rs2229774, SLC28A3 rs7853758 and UGT1A6 rs17863783 variants currently have the strongest and the most consistent evidence for association with ACT. Genetic variants in ABCC1, ABCC2, ABCC5, ABCB1, ABCB4, CBR3, RAC2, NCF4, CYBA, GSTP1, CAT, SULT2B1, POR, HAS3, SLC22A7, SCL22A17, HFE and NOS3 have also been associated with ACT, but require additional validation. We recommend pharmacogenomic testing for the RARG rs2229774 (S427L), SLC28A3 rs7853758 (L461L) and UGT1A6*4 rs17863783 (V209V) variants in childhood cancer patients with an indication for doxorubicin or daunorubicin therapy (Level B - moderate). Based on an overall risk stratification, taking into account genetic and clinical risk factors, we recommend a number of management options including increased frequency of echocardiogram monitoring, follow-up, as well as therapeutic options within the current standard of clinical practice. CONCLUSIONS Existing evidence demonstrates that genetic factors have the potential to improve the discrimination between individuals at higher and lower risk of ACT. Genetic testing may therefore support both patient care decisions and evidence development for an improved prevention of ACT.
Collapse
Affiliation(s)
- Folefac Aminkeng
- Centre for Molecular Medicine and Therapeutics, Department of Medical GeneticsUniversity of British ColumbiaVancouverBCCanada
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Colin J. D. Ross
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Shahrad R. Rassekh
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Pediatric Hematology/Oncology/BMT, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Soomi Hwang
- Faculty of Pharmaceutical SciencesUniversity of British ColumbiaVancouverBCCanada
| | | | - Amit P. Bhavsar
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Anne Smith
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Pharmaceutical Outcomes & Policy Innovations ProgrammeBC Children's HospitalVancouverBCCanada
| | - Shubhayan Sanatani
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
| | | | - Daniel Bernstein
- Department of Pediatrics, Division of CardiologyStanford UniversityStanfordCAUSA
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, Department of Medical GeneticsUniversity of British ColumbiaVancouverBCCanada
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Translational Laboratory in Genetic Medicine, National University of Singapore and Association for ScienceTechnology and Research (A*STAR)Singapore
| | - Ursula Amstutz
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital and University of BernSwitzerland
| | - Bruce C. Carleton
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Pharmaceutical Outcomes & Policy Innovations ProgrammeBC Children's HospitalVancouverBCCanada
| | | |
Collapse
|
30
|
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
|
31
|
Bettadapur A, Suh GC, Geisse NA, Wang ER, Hua C, Huber HA, Viscio AA, Kim JY, Strickland JB, McCain ML. Prolonged Culture of Aligned Skeletal Myotubes on Micromolded Gelatin Hydrogels. Sci Rep 2016; 6:28855. [PMID: 27350122 PMCID: PMC4924097 DOI: 10.1038/srep28855] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/10/2016] [Indexed: 12/19/2022] Open
Abstract
In vitro models of skeletal muscle are critically needed to elucidate disease mechanisms, identify therapeutic targets, and test drugs pre-clinically. However, culturing skeletal muscle has been challenging due to myotube delamination from synthetic culture substrates approximately one week after initiating differentiation from myoblasts. In this study, we successfully maintained aligned skeletal myotubes differentiated from C2C12 mouse skeletal myoblasts for three weeks by utilizing micromolded (μmolded) gelatin hydrogels as culture substrates, which we thoroughly characterized using atomic force microscopy (AFM). Compared to polydimethylsiloxane (PDMS) microcontact printed (μprinted) with fibronectin (FN), cell adhesion on gelatin hydrogel constructs was significantly higher one week and three weeks after initiating differentiation. Delamination from FN-μprinted PDMS precluded robust detection of myotubes. Compared to a softer blend of PDMS μprinted with FN, myogenic index, myotube width, and myotube length on μmolded gelatin hydrogels was similar one week after initiating differentiation. However, three weeks after initiating differentiation, these parameters were significantly higher on μmolded gelatin hydrogels compared to FN-μprinted soft PDMS constructs. Similar results were observed on isotropic versions of each substrate, suggesting that these findings are independent of substrate patterning. Our platform enables novel studies into skeletal muscle development and disease and chronic drug testing in vitro.
Collapse
Affiliation(s)
- Archana Bettadapur
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Gio C Suh
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | | | - Evelyn R Wang
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.,Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Clara Hua
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Holly A Huber
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Alyssa A Viscio
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Joon Young Kim
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Julie B Strickland
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
32
|
Bagchi RA, Lin J, Wang R, Czubryt MP. Regulation of fibronectin gene expression in cardiac fibroblasts by scleraxis. Cell Tissue Res 2016; 366:381-391. [PMID: 27324126 DOI: 10.1007/s00441-016-2439-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
Abstract
The glycoprotein fibronectin is a key component of the extracellular matrix. By interacting with numerous matrix and cell surface proteins, fibronectin plays important roles in cell adhesion, migration and intracellular signaling. Up-regulation of fibronectin occurs in tissue fibrosis, and previous studies have identified the pro-fibrotic factor TGFβ as an inducer of fibronectin expression, although the mechanism responsible remains unknown. We have previously shown that a key downstream effector of TGFβ signaling in cardiac fibroblasts is the transcription factor scleraxis, which in turn regulates the expression of a wide variety of extracellular matrix genes. We noted that fibronectin expression tracked closely with scleraxis expression, but it was unclear whether scleraxis directly regulated the fibronectin gene. Here, we report that scleraxis acts via two E-box binding sites in the proximal human fibronectin promoter to govern fibronectin expression, with the second E-box being both sufficient and necessary for scleraxis-mediated fibronectin expression to occur. A combination of electrophoretic mobility shift and chromatin immunoprecipitation assays indicated that scleraxis interacted to a greater degree with the second E-box. Over-expression or knockdown of scleraxis resulted in increased or decreased fibronectin expression, respectively, and scleraxis null mice presented with dramatically decreased immunolabeling for fibronectin in cardiac tissue sections compared to wild-type controls. Furthermore, scleraxis was required for TGFβ-induced fibronectin expression: TGFβ lost its ability to induce fibronectin expression following scleraxis knockdown. Together, these results demonstrate a novel and required role for scleraxis in the regulation of cardiac fibroblast fibronectin gene expression basally or in response to TGFβ.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada.,Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Division of Cardiology, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, RC2- Room 8450, Aurora, CO, 80045, USA
| | - Justin Lin
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Ryan Wang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Michael P Czubryt
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
33
|
Korompelis P, Piperi C, Adamopoulos C, Dalagiorgou G, Korkolopoulou P, Sepsa A, Antsaklis A, Papavassiliou AG. Expression of vascular endothelial factor-A, gelatinases (MMP-2, MMP-9) and TIMP-1 in uterine leiomyomas. Clin Chem Lab Med 2016; 53:1415-24. [PMID: 25470608 DOI: 10.1515/cclm-2014-0798] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND Leiomyomas growth involves cellular hypertrophy, modulation of mitotic activity and upregulation of extracellular matrix (ECM). Vascular factors and matrix metalloproteinases (MMPs) play a coordinated role during neoplasia and tissue remodeling. The present study investigates the role of angiogenic factor vascular endothelial growth factor (VEGF)-A with the activity of main gelatinases, MMP-2/MMP-9 and their tissue inhibitor TIMP-1 in patients with leiomyomas. METHODS Peripheral blood of 46 women with uterine leiomyomas was obtained prior hysterectomy to assess VEGF-A, MMP-2, -9, TIMP-1 levels by enzyme-linked immunosorbent assay compared to 39 healthy controls. Protein expression levels of VEGF-A, MMP-2 and MMP-9 were evaluated by western immunoblotting and immunohistochemistry in leiomyomas tissue specimens after hysterectomy. Furthermore, the activity of gelatinases in leiomyoma tissue extracts and control myometrium was evaluated by semi-quantitative zymography. RESULTS Circulating levels of VEGF-A, MMP-2 and TIMP-1 were significantly elevated in leiomyoma patients compared to controls (p<0.001, p=0.004, p=0.003, respectively). A positive correlation was found between VEGF-A and MMP-2 (p=0.021) as well as MMP-9 (p=0.001) peripheral levels in the patient's group. Furthermore, increased VEGF-A protein levels were detected in leiomyoma tissue compared to control myometrium, followed by increased localization of both VEGF-A and MMP-2 in the ECM embedding bundles of smooth muscle cells of leiomyomas. The activity of MMP-2 was significantly higher in leiomyomas than normal myometrium in all investigated tissues. CONCLUSIONS This study demonstrates a possible coordinated role of VEGF-A and MMP-2 during uterine leiomyomas growth and angiogenesis with potential prognostic significance.
Collapse
|
34
|
Xu Y, Guan J. Biomaterial property-controlled stem cell fates for cardiac regeneration. Bioact Mater 2016; 1:18-28. [PMID: 29744392 PMCID: PMC5883968 DOI: 10.1016/j.bioactmat.2016.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction (MI) affects more than 8 million people in the United States alone. Due to the insufficient regeneration capacity of the native myocardium, one widely studied approach is cardiac tissue engineering, in which cells are delivered with or without biomaterials and/or regulatory factors to fully regenerate the cardiac functions. Specifically, in vitro cardiac tissue engineering focuses on using biomaterials as a reservoir for cells to attach, as well as a carrier of various regulatory factors such as growth factors and peptides, providing high cell retention and a proper microenvironment for cells to migrate, grow and differentiate within the scaffolds before implantation. Many studies have shown that the full establishment of a functional cardiac tissue in vitro requires synergistic actions between the seeded cells, the tissue culture condition, and the biochemical and biophysical environment provided by the biomaterials-based scaffolds. Proper electrical stimulation and mechanical stretch during the in vitro culture can induce the ordered orientation and differentiation of the seeded cells. On the other hand, the various scaffolds biochemical and biophysical properties such as polymer composition, ligand concentration, biodegradability, scaffold topography and mechanical properties can also have a significant effect on the cellular processes. Cell therapy is an attractive approach for cardiac regeneration after myocardial infarction. Biomaterials are used as cell carriers. This review highlights how biochemical and biophysical properties of biomaterials affect cell fates.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
35
|
Stem cells and injectable hydrogels: Synergistic therapeutics in myocardial repair. Biotechnol Adv 2016; 34:362-379. [PMID: 26976812 DOI: 10.1016/j.biotechadv.2016.03.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 02/27/2016] [Accepted: 03/07/2016] [Indexed: 02/08/2023]
Abstract
One of the major problems in the treatment of cardiovascular diseases is the inability of myocardium to self-regenerate. Current therapies are unable to restore the heart's function after myocardial infarction. Myocardial tissue engineering is potentially a key approach to regenerate damaged heart muscle. Myocardial patches are applied surgically, whereas injectable hydrogels provide effective minimally invasive approaches to recover functional myocardium. These hydrogels are easily administered and can be either cell free or loaded with bioactive agents and/or cardiac stem cells, which may apply paracrine effects. The aim of this review is to investigate the advantages and disadvantages of injectable stem cell-laden hydrogels and highlight their potential applications for myocardium repair.
Collapse
|
36
|
Extracellular matrix-mediated cellular communication in the heart. J Mol Cell Cardiol 2016; 91:228-37. [PMID: 26778458 DOI: 10.1016/j.yjmcc.2016.01.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.
Collapse
|
37
|
Padmanabhan Iyer R, Chiao YA, Flynn ER, Hakala K, Cates CA, Weintraub ST, de Castro Brás LE. Matrix metalloproteinase-9-dependent mechanisms of reduced contractility and increased stiffness in the aging heart. Proteomics Clin Appl 2015; 10:92-107. [PMID: 26415707 DOI: 10.1002/prca.201500038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/12/2015] [Accepted: 09/22/2015] [Indexed: 12/23/2022]
Abstract
PURPOSE Matrix metalloproteinases (MMPs) collectively degrade all extracellular matrix (ECM) proteins. Of the MMPs, MMP-9 has the strongest link to the development of cardiac dysfunction. Aging associates with increased MMP-9 expression in the left ventricle (LV) and reduced cardiac function. We investigated the effect of MMP-9 deletion on the cardiac ECM in aged animals. EXPERIMENTAL DESIGN We used male and female middle-aged (10- to16-month old) and old (20- to 24-month old) wild-type (WT) and MMP-9 null mice (n = 6/genotype/age). LVs were decellularized to remove highly abundant mitochondrial proteins that could mask identification of relative lower abundant components, analyzed by shotgun proteomics, and proteins of interest validated by immunoblot. RESULTS Elastin microfibril interface-located protein 1 (EMILIN-1) decreased with age in WT (p < 0.05), but not in MMP-9 null. EMILIN-1 promotes integrin-dependent cell adhesion and EMILIN-1 deficiency has been associated with vascular stiffening. Talin-2, a cytoskeletal protein, was elevated with age in WT (p < 0.05), and MMP-9 deficiency blunted this increase. Talin-2 is highly expressed in adult cardiac myocytes, transduces mechanical force to the ECM, and is activated by increases in substrate stiffness. Our results suggest that MMP-9 deletion may reduce age-related myocardial stiffness, which may explain improved cardiac function in MMP-9 null animals. CONCLUSIONS We identified age-related changes in the cardiac proteome that are MMP-9 dependent, suggesting MMP-9 as a possible therapeutic target for the aging patient.
Collapse
Affiliation(s)
- Rugmani Padmanabhan Iyer
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Physiology and Biophysics, Mississippi Center for Heart Research, Jackson, MS, USA
| | - Ying Ann Chiao
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Elizabeth R Flynn
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Physiology and Biophysics, Mississippi Center for Heart Research, Jackson, MS, USA
| | - Kevin Hakala
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Courtney A Cates
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Physiology and Biophysics, Mississippi Center for Heart Research, Jackson, MS, USA
| | - Susan T Weintraub
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lisandra E de Castro Brás
- San Antonio Cardiovascular Proteomics Center, San Antonio, TX, USA.,Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| |
Collapse
|
38
|
Embryonic Lethality Due to Arrested Cardiac Development in Psip1/Hdgfrp2 Double-Deficient Mice. PLoS One 2015; 10:e0137797. [PMID: 26367869 PMCID: PMC4569352 DOI: 10.1371/journal.pone.0137797] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022] Open
Abstract
Hepatoma-derived growth factor (HDGF) related protein 2 (HRP2) and lens epithelium-derived growth factor (LEDGF)/p75 are closely related members of the HRP2 protein family. LEDGF/p75 has been implicated in numerous human pathologies including cancer, autoimmunity, and infectious disease. Knockout of the Psip1 gene, which encodes for LEDGF/p75 and the shorter LEDGF/p52 isoform, was previously shown to cause perinatal lethality in mice. The function of HRP2 was by contrast largely unknown. To learn about the role of HRP2 in development, we knocked out the Hdgfrp2 gene, which encodes for HRP2, in both normal and Psip1 knockout mice. Hdgfrp2 knockout mice developed normally and were fertile. By contrast, the double deficient mice died at approximate embryonic day (E) 13.5. Histological examination revealed ventricular septal defect (VSD) associated with E14.5 double knockout embryos. To investigate the underlying molecular mechanism(s), RNA recovered from ventricular tissue was subjected to RNA-sequencing on the Illumina platform. Bioinformatic analysis revealed several genes and biological pathways that were significantly deregulated by the Psip1 knockout and/or Psip1/Hdgfrp2 double knockout. Among the dozen genes known to encode for LEDGF/p75 binding factors, only the expression of Nova1, which encodes an RNA splicing factor, was significantly deregulated by the knockouts. However the expression of other RNA splicing factors, including the LEDGF/p52-interacting protein ASF/SF2, was not significantly altered, indicating that deregulation of global RNA splicing was not a driving factor in the pathology of the VSD. Tumor growth factor (Tgf) β-signaling, which plays a key role in cardiac morphogenesis during development, was the only pathway significantly deregulated by the double knockout as compared to control and Psip1 knockout samples. We accordingly speculate that deregulated Tgf-β signaling was a contributing factor to the VSD and prenatal lethality of Psip1/Hdgfrp2 double-deficient mice.
Collapse
|
39
|
Rosellini E, Cristallini C, Guerra GD, Barbani N. Surface chemical immobilization of bioactive peptides on synthetic polymers for cardiac tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:515-33. [PMID: 25787756 DOI: 10.1080/09205063.2015.1030991] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The aim of this work was the development of new synthetic polymeric systems, functionalized by surface chemical modification with bioactive peptides, for myocardial tissue engineering. Polycaprolactone and a poly(ester-ether-ester) block copolymer synthesized in our lab, polycaprolactone-poly(ethylene oxide)-polycaprolactone (PCL-PEO-PCL), were used as the substrates to be modified. Two pentapeptides, H-Gly-Arg-Gly-Asp-Ser-OH (GRGDS) from fibronectin and H-Tyr-Ile-Gly-Ser-Arg-OH (YIGSR) from laminin, were used for the functionalization. Polymeric membranes were obtained by casting from solutions and then functionalized by means of alkaline hydrolysis and subsequent coupling of the bioactive molecules through 1-(3-dimethylaminopropyl)-3-ethylcarbodimide hydrochloride/N-hydroxysuccinimide chemistry. The hydrolysis conditions, in terms of hydrolysis time, temperature, and sodium hydroxide concentration, were optimized for the two materials. The occurrence of the coupling reaction was demonstrated by infrared spectroscopy, as the presence on the functionalized materials of the absorption peaks typical of the two peptides. The peptide surface density was determined by chromatographic analysis and the distribution was studied by infrared chemical imaging. The results showed a nearly homogeneous peptide distribution, with a density above the minimum value necessary to promote cell adhesion. Preliminary in vitro cell culture studies demonstrated that the introduction of the bioactive molecules had a positive effect on improving C2C12 myoblasts growth on the synthetic materials.
Collapse
Affiliation(s)
- Elisabetta Rosellini
- a Department of Civil and Industrial Engineering , University of Pisa , Largo Lucio Lazzarino, 56126 Pisa , Italy
| | | | | | | |
Collapse
|
40
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Dostal D, Glaser S, Baudino TA. Cardiac Fibroblast Physiology and Pathology. Compr Physiol 2015; 5:887-909. [DOI: 10.1002/cphy.c140053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
42
|
Methe K, Bäckdahl H, Johansson BR, Nayakawde N, Dellgren G, Sumitran-Holgersson S. An alternative approach to decellularize whole porcine heart. Biores Open Access 2014; 3:327-38. [PMID: 25469317 PMCID: PMC4245870 DOI: 10.1089/biores.2014.0046] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Scaffold characteristics are decisive for repopulating the acellular tissue with cells. A method to produce such a scaffold from intact organ requires a customized decellularization protocol. Here, we have decellularized whole, intact porcine hearts by serial perfusion and agitation of hypotonic solution, an ionic detergent (4% sodium deoxycholate), and a nonionic detergent (1% Triton X-100). The resultant matrix was characterized for its degree of decellularization, morphological and functional integrity. The protocol used resulted in extensive decellularization of the cardiac tissue, but the cytoskeletal elements (contractile apparatus) of cardiomyocytes remained largely unaffected by the procedure although their membranous organelles were completely absent. Further, several residual angiogenic growth factors were found to be present in the decellularized tissue.
Collapse
Affiliation(s)
- Ketaki Methe
- Transplantation Institute and Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Bäckdahl
- Department of Chemistry and Materials, SP Technical Research Institute of Sweden, Borås, Sweden
| | - Bengt R. Johansson
- The Electron Microscopy Unit, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nikhil Nayakawde
- Transplantation Institute and Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Goran Dellgren
- Transplant Institute and Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Suchitra Sumitran-Holgersson
- Transplantation Institute and Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
43
|
Di Vito A, Mignogna C, Donato G. The mysterious pathways of cardiac myxomas: a review of histogenesis, pathogenesis and pathology. Histopathology 2014; 66:321-32. [DOI: 10.1111/his.12531] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Anna Di Vito
- Department of Clinical and Experimental Medicine; University of Catanzaro ‘Magna Graecia Medical School; Catanzaro Italy
| | - Chiara Mignogna
- Department of Health Science, Pathology Unit; University of Catanzaro ‘Magna Graecia Medical School; Catanzaro Italy
| | - Giuseppe Donato
- Department of Health Science, Pathology Unit; University of Catanzaro ‘Magna Graecia Medical School; Catanzaro Italy
| |
Collapse
|
44
|
Zhu R, Blazeski A, Poon E, Costa KD, Tung L, Boheler KR. Physical developmental cues for the maturation of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2014; 5:117. [PMID: 25688759 PMCID: PMC4396914 DOI: 10.1186/scrt507] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are the most promising source of cardiomyocytes (CMs) for experimental and clinical applications, but their use is largely limited by a structurally and functionally immature phenotype that most closely resembles embryonic or fetal heart cells. The application of physical stimuli to influence hPSC-CMs through mechanical and bioelectrical transduction offers a powerful strategy for promoting more developmentally mature CMs. Here we summarize the major events associated with in vivo heart maturation and structural development. We then review the developmental state of in vitro derived hPSC-CMs, while focusing on physical (electrical and mechanical) stimuli and contributory (metabolic and hypertrophic) factors that are actively involved in structural and functional adaptations of hPSC-CMs. Finally, we highlight areas for possible future investigation that should provide a better understanding of how physical stimuli may promote in vitro development and lead to mechanistic insights. Advances in the use of physical stimuli to promote developmental maturation will be required to overcome current limitations and significantly advance research of hPSC-CMs for cardiac disease modeling, in vitro drug screening, cardiotoxicity analysis and therapeutic applications.
Collapse
|
45
|
Liu Z, Li W, Ma X, Ding N, Spallotta F, Southon E, Tessarollo L, Gaetano C, Mukouyama YS, Thiele CJ. Essential role of the zinc finger transcription factor Casz1 for mammalian cardiac morphogenesis and development. J Biol Chem 2014; 289:29801-16. [PMID: 25190801 DOI: 10.1074/jbc.m114.570416] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chromosome 1p36 deletion syndrome is one of the most common terminal deletions observed in humans and is related to congenital heart disease (CHD). However, the 1p36 genes that contribute to heart disease have not been clearly delineated. Human CASZ1 gene localizes to 1p36 and encodes a zinc finger transcription factor. Casz1 is required for Xenopus heart ventral midline progenitor cell differentiation. Whether Casz1 plays a role during mammalian heart development is unknown. Our aim is to determine 1p36 gene CASZ1 function at regulating heart development in mammals. We generated a Casz1 knock-out mouse using Casz1-trapped embryonic stem cells. Casz1 deletion in mice resulted in abnormal heart development including hypoplasia of myocardium, ventricular septal defect, and disorganized morphology. Hypoplasia of myocardium was caused by decreased cardiomyocyte proliferation. Comparative genome-wide RNA transcriptome analysis of Casz1 depleted embryonic hearts identifies abnormal expression of genes that are critical for muscular system development and function, such as muscle contraction genes TNNI2, TNNT1, and CKM; contractile fiber gene ACTA1; and cardiac arrhythmia associated ion channel coding genes ABCC9 and CACNA1D. The transcriptional regulation of some of these genes by Casz1 was also found in cellular models. Our results showed that loss of Casz1 during mouse development led to heart defect including cardiac noncompaction and ventricular septal defect, which phenocopies 1p36 deletion syndrome related CHD. This suggests that CASZ1 is a novel 1p36 CHD gene and that the abnormal expression of cardiac morphogenesis and contraction genes induced by loss of Casz1 contributes to the heart defect.
Collapse
Affiliation(s)
| | - Wenling Li
- the Laboratories of Stem Cell and Neuro-vascular Biology and
| | - Xuefei Ma
- the Molecular Cardiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892, and
| | | | - Francesco Spallotta
- the Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany
| | - Eileen Southon
- the Mouse Cancer Genetics Program, Neural Development Section, National Cancer Institute, Bethesda, Maryland 20892
| | - Lino Tessarollo
- the Mouse Cancer Genetics Program, Neural Development Section, National Cancer Institute, Bethesda, Maryland 20892
| | - Carlo Gaetano
- the Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany
| | | | | |
Collapse
|
46
|
Sa S, Wong L, McCloskey KE. Combinatorial fibronectin and laminin signaling promote highly efficient cardiac differentiation of human embryonic stem cells. Biores Open Access 2014; 3:150-61. [PMID: 25126479 PMCID: PMC4120929 DOI: 10.1089/biores.2014.0018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiomyocytes (CMs) differentiated from human embryonic stem cells (hESCs) are a promising and potentially unlimited cell source for myocardial repair and regeneration. Recently, multiple methodologies-primarily based on the optimization of growth factors-have been described for efficient cardiac differentiation of hESCs. However, the role of extracellular matrix (ECM) signaling in CM differentiation has not yet been explored fully. This study examined the role of ECM signaling in the efficient generation of CMs from both H7 and H9 ESCs. The hESCs were differentiated on ECM substrates composed of a range of fibronectin (FN) and laminin (LN) ratios and gelatin and evaluated by the fluorescence activated cell scanning (FACS) analysis on day 14. Of the ECM substrates examined, the 70:30 FN:LN reproducibly generated the greatest numbers of CMs from both hESC lines. Moreover, the LN receptor integrin β4 (ITGB4) and FN receptor integrin β5 (ITGB5) genes, jointly with increased phosphorylated focal adhension kinase and phosphorylated extracellular signal-regulated kinases (p-ERKs), were up-regulated over 13-fold in H7 and H9 cultured on 70:30 FN:LN compared with gelatin. Blocking studies confirmed the role of all these molecules in CM specification, suggesting that the 70:30 FN:LN ECM promotes highly efficient differentiation of CMs through the integrin-mediated MEK/ERK signaling pathway. Lastly, the data suggest that FN:LN-induced signaling utilizes direct cell-to-cell signaling from distinct ITGB4(+) and ITGB5(+) cells.
Collapse
Affiliation(s)
- Silin Sa
- Graduate Group in Biological Engineering & Small-Scale Technologies, University of California, Merced, California
| | - Lian Wong
- Graduate Group in Biological Engineering & Small-Scale Technologies, University of California, Merced, California
| | - Kara E. McCloskey
- Graduate Group in Biological Engineering & Small-Scale Technologies, University of California, Merced, California
- School of Engineering, University of California, Merced, California
| |
Collapse
|
47
|
A Novel Seeding and Conditioning Bioreactor for Vascular Tissue Engineering. Processes (Basel) 2014. [DOI: 10.3390/pr2030526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
McCain ML, Agarwal A, Nesmith HW, Nesmith AP, Parker KK. Micromolded gelatin hydrogels for extended culture of engineered cardiac tissues. Biomaterials 2014; 35:5462-71. [PMID: 24731714 DOI: 10.1016/j.biomaterials.2014.03.052] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/21/2014] [Indexed: 12/12/2022]
Abstract
Defining the chronic cardiotoxic effects of drugs during preclinical screening is hindered by the relatively short lifetime of functional cardiac tissues in vitro, which are traditionally cultured on synthetic materials that do not recapitulate the cardiac microenvironment. Because collagen is the primary extracellular matrix protein in the heart, we hypothesized that micromolded gelatin hydrogel substrates tuned to mimic the elastic modulus of the heart would extend the lifetime of engineered cardiac tissues by better matching the native chemical and mechanical microenvironment. To measure tissue stress, we used tape casting, micromolding, and laser engraving to fabricate gelatin hydrogel muscular thin film cantilevers. Neonatal rat cardiac myocytes adhered to gelatin hydrogels and formed aligned tissues as defined by the microgrooves. Cardiac tissues could be cultured for over three weeks without declines in contractile stress. Myocytes on gelatin had higher spare respiratory capacity compared to those on fibronectin-coated PDMS, suggesting that improved metabolic function could be contributing to extended culture lifetime. Lastly, human induced pluripotent stem cell-derived cardiac myocytes adhered to micromolded gelatin surfaces and formed aligned tissues that remained functional for four weeks, highlighting their potential for human-relevant chronic studies.
Collapse
Affiliation(s)
- Megan L McCain
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Ashutosh Agarwal
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Haley W Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Alexander P Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
49
|
Age and SPARC change the extracellular matrix composition of the left ventricle. BIOMED RESEARCH INTERNATIONAL 2014; 2014:810562. [PMID: 24783223 PMCID: PMC3982264 DOI: 10.1155/2014/810562] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 01/08/2023]
Abstract
Secreted protein acidic and rich in cysteine (SPARC), a collagen-binding matricellular protein, has been implicated in procollagen processing and deposition. The aim of this study was to investigate age- and SPARC-dependent changes in protein composition of the cardiac extracellular matrix (ECM). We studied 6 groups of mice (n = 4/group): young (4-5 months old), middle-aged (11-12 m.o.), and old (18–29 m.o.) C57BL/6J wild type (WT) and SPARC null. The left ventricle (LV) was decellularized to enrich for ECM proteins. Protein extracts were separated by SDS-PAGE, digested in-gel, and analyzed by HPLC-ESI-MS/MS. Relative quantification was performed by spectral counting, and changes in specific proteins were validated by immunoblotting. We identified 321 proteins, of which 44 proteins were extracellular proteins. Of these proteins, collagen III levels were lower in the old null mice compared to WT, suggestive of a role for SPARC in collagen deposition. Additionally, fibrillin showed a significant increase in the null middle-aged group, suggestive of increased microfibril deposition in the absence of SPARC. Collagen VI increased with age in both genotypes (>3-fold), while collagen IV showed increased age-associated levels only in the WT animals (4-fold, P < 0.05). These changes may explain the previously reported age-associated increases in LV stiffness. In summary, our data suggest SPARC is a possible therapeutic target for aging induced LV dysfunction.
Collapse
|
50
|
Hazeltine LB, Badur MG, Lian X, Das A, Han W, Palecek SP. Temporal impact of substrate mechanics on differentiation of human embryonic stem cells to cardiomyocytes. Acta Biomater 2014; 10:604-12. [PMID: 24200714 PMCID: PMC3889126 DOI: 10.1016/j.actbio.2013.10.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 10/16/2013] [Accepted: 10/28/2013] [Indexed: 12/13/2022]
Abstract
A significant clinical need exists to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes, enabling tissue modeling for in vitro discovery of new drugs or cell-based therapies for heart repair in vivo. Chemical and mechanical microenvironmental factors are known to impact the efficiency of stem cell differentiation, but cardiac differentiation protocols in hPSCs are typically performed on rigid tissue culture polystyrene (TCPS) surfaces, which do not present a physiological mechanical setting. To investigate the temporal effects of mechanics on cardiac differentiation, we cultured human embryonic stem cells (hESCs) and their derivatives on polyacrylamide hydrogel substrates with a physiologically relevant range of stiffnesses. In directed differentiation and embryoid body culture systems, differentiation of hESCs to cardiac troponin T-expressing (cTnT+) cardiomyocytes peaked on hydrogels of intermediate stiffness. Brachyury expression also peaked on intermediate stiffness hydrogels at day 1 of directed differentiation, suggesting that stiffness impacted the initial differentiation trajectory of hESCs to mesendoderm. To investigate the impact of substrate mechanics during cardiac specification of mesodermal progenitors, we initiated directed cardiomyocyte differentiation on TCPS and transferred cells to hydrogels at the Nkx2.5/Isl1+ cardiac progenitor cell stage. No differences in cardiomyocyte purity with stiffness were observed on day 15. These experiments indicate that differentiation of hESCs is sensitive to substrate mechanics at early stages of mesodermal induction, and proper application of substrate mechanics can increase the propensity of hESCs to differentiate to cardiomyocytes.
Collapse
Affiliation(s)
- Laurie B Hazeltine
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mehmet G Badur
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaojun Lian
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amritava Das
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wenqing Han
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|