279 results on '"Heart Valves cytology"'
Search Results
2. The Jagged-1/Notch1 Signaling Pathway Promotes the Construction of Tissue-Engineered Heart Valves.
- Author
-
Liu K, Wei ZY, Zhong XH, Liu X, Chen H, Pan Y, and Zeng W
- Subjects
- Animals, Humans, Epithelial-Mesenchymal Transition drug effects, Heart Valves cytology, Heart Valves metabolism, Swine, Cell Proliferation drug effects, Tissue Scaffolds chemistry, Heart Valve Prosthesis, Endothelial Cells metabolism, Endothelial Cells cytology, Endothelial Cells drug effects, Signal Transduction drug effects, Tissue Engineering methods, Jagged-1 Protein metabolism, Receptor, Notch1 metabolism
- Abstract
Background: Tissue-engineered heart valves (TEHVs) are promising new heart valve substitutes for valvular heart disease. The Notch signaling pathway plays a critical role in the development of congenital heart valves. Objective: To investigate the role of the Notch signaling pathway in the construction of TEHVs. Methods: The induced endothelial cells, which act as seed cells, were differentiated from adipose-derived stem cells and were treated with Jagged-1 (JAG-1) protein and γ-secretase inhibitor (DAPT, N -[ N -(3,5-difluorophenacetyl)-l-alanyl]-s-phenylglycine t-butyl ester), respectively. Cell phenotypic changes, the expression of proteins relating to the epithelial-mesenchymal transition (EMT), and changes in paxillin expression were detected. Decellularized valve scaffolds were produced from decellularized porcine aortic valves. The seed cells were them inoculated into Matrigel-coated flap scaffolds for complex culture and characterization. Results: JAG-1 significantly reduced apoptosis and promoted the seeded cells' proliferation and migration ability, in contrast to the treatment of DAPT. In addition, the expression of EMT-related proteins, E-cadherin and N-cadherin, was significantly increased after treatment with JAG-1 and was reduced after the application of DAPT. Meanwhile, the adhesive-related expression of paxillin and fibronectin proteins was increased after the activation of Notch1 signaling and vice versa. Of interest, activation of the Notch1 signaling pathway resulted in more closely arranged cells on the valve surface after recellularization. Conclusion: Activation of the JAG-1/Notch1 signaling pathway increased seeded cells' proliferation and migratory ability and promoted the EMT and adhesion of seed cells, which was conducive to binding to the matrix, facilitating accelerated endothelialization of TEHVs.
- Published
- 2024
- Full Text
- View/download PDF
3. Directed Differentiation of Human Induced Pluripotent Stem Cells to Heart Valve Cells.
- Author
-
Cai Z, Zhu M, Xu L, Wang Y, Xu Y, Yim WY, Cao H, Guo R, Qiu X, He X, Shi J, Qiao W, and Dong N
- Subjects
- Humans, Cells, Cultured, Endothelial Cells metabolism, Endothelial Cells cytology, Signal Transduction, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Cell Differentiation, Heart Valves cytology, Heart Valves metabolism
- Abstract
Background: A main obstacle in current valvular heart disease research is the lack of high-quality homogeneous functional heart valve cells. Human induced pluripotent stem cells (hiPSCs)-derived heart valve cells may help with this dilemma. However, there are no well-established protocols to induce hiPSCs to differentiate into functional heart valve cells, and the networks that mediate the differentiation have not been fully elucidated., Methods: To generate heart valve cells from hiPSCs, we sequentially activated the Wnt, BMP4, VEGF (vascular endothelial growth factor), and NFATc1 signaling pathways using CHIR-99021, BMP4, VEGF-165, and forskolin, respectively. The transcriptional and functional similarity of hiPSC-derived heart valve cells compared with primary heart valve cells were characterized. Longitudinal single-cell RNA sequencing was used to uncover the trajectory, switch genes, pathways, and transcription factors of the differentiation., Results: An efficient protocol was developed to induce hiPSCs to differentiate into functional hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells. After 6-day differentiation and CD144 magnetic bead sorting, ≈70% CD144
+ cells and 30% CD144- cells were obtained. On the basis of single-cell RNA sequencing data, the CD144+ cells and CD144- cells were found to be highly similar to primary heart valve endothelial cells and primary heart valve interstitial cells in gene expression profile. Furthermore, CD144+ cells had the typical function of primary heart valve endothelial cells, including tube formation, uptake of low-density lipoprotein, generation of endothelial nitric oxide synthase, and response to shear stress. Meanwhile, CD144- cells could secret collagen and matrix metalloproteinases, and differentiate into osteogenic or adipogenic lineages like primary heart valve interstitial cells. Therefore, we identified CD144+ cells and CD144- cells as hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells, respectively. Using single-cell RNA sequencing analysis, we demonstrated that the trajectory of heart valve cell differentiation was consistent with embryonic valve development. We identified the main switch genes (NOTCH1, HEY1, and MEF2C), signaling pathways (TGF-β, Wnt, and NOTCH), and transcription factors (MSX1, SP5, and MECOM) that mediated the differentiation. Finally, we found that hiPSC-derived valve interstitial-like cells might derive from hiPSC-derived valve endothelial-like cells undergoing endocardial-mesenchymal transition., Conclusions: In summary, this is the first study to report an efficient strategy to generate functional hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells from hiPSCs, as well as to elucidate the differentiation trajectory and transcriptional dynamics of hiPSCs differentiated into heart valve cells., Competing Interests: Disclosures None.- Published
- 2024
- Full Text
- View/download PDF
4. Valvular Endothelial Cell Response to the Mechanical Environment-A Review.
- Author
-
Deb N and Lacerda CMR
- Subjects
- Humans, Animals, Extracellular Matrix metabolism, Signal Transduction, Mechanotransduction, Cellular, Endothelial Cells cytology, Endothelial Cells physiology, Heart Valves cytology, Heart Valves physiology
- Abstract
Heart valve leaflets are complex structures containing valve endothelial cells, interstitial cells, and extracellular matrix. Heart valve endothelial cells sense mechanical stimuli, and communicate amongst themselves and the surrounding cells and extracellular matrix to maintain tissue homeostasis. In the presence of abnormal mechanical stimuli, endothelial cell communication is triggered in defense and such processes may eventually lead to cardiac disease progression. This review focuses on the role of mechanical stimuli on heart valve endothelial surfaces-from heart valve development and maintenance of tissue integrity to disease progression with related signal pathways involved in this process., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
5. Bioelectric signaling and the control of cardiac cell identity in response to mechanical forces.
- Author
-
Fukui H, Chow RW, Xie J, Foo YY, Yap CH, Minc N, Mochizuki N, and Vermot J
- Subjects
- Adenosine Triphosphate metabolism, Animals, Calcium metabolism, Calcium Signaling, Electrophysiological Phenomena, Endothelial Cells physiology, Heart Valves cytology, Heart Valves metabolism, NFATC Transcription Factors metabolism, Receptors, Purinergic P2 metabolism, Zebrafish, Heart Valves growth & development, Shear Strength, Stress, Mechanical
- Abstract
Developing cardiovascular systems use mechanical forces to take shape, but how ubiquitous blood flow forces instruct local cardiac cell identity is still unclear. By manipulating mechanical forces in vivo, we show here that shear stress is necessary and sufficient to promote valvulogenesis. We found that valve formation is associated with the activation of an extracellular adenosine triphosphate (ATP)–dependent purinergic receptor pathway, specifically triggering calcium ion (Ca
2+ ) pulses and nuclear factor of activated T cells 1 (Nfatc1) activation. Thus, mechanical forces are converted into discrete bioelectric signals by an ATP-Ca2+ -Nfatc1–mechanosensitive pathway to generate positional information and control valve formation.- Published
- 2021
- Full Text
- View/download PDF
6. Generation and characterization of cardiac valve endothelial-like cells from human pluripotent stem cells.
- Author
-
Cheng L, Xie M, Qiao W, Song Y, Zhang Y, Geng Y, Xu W, Wang L, Wang Z, Huang K, Dong N, and Sun Y
- Subjects
- Animals, Humans, Sus scrofa, Bone Morphogenetic Protein 4 administration & dosage, Endothelial Cells cytology, Heart Valves cytology, Pluripotent Stem Cells cytology, Transforming Growth Factor beta1 administration & dosage
- Abstract
The cardiac valvular endothelial cells (VECs) are an ideal cell source that could be used for making the valve organoids. However, few studies have been focused on the derivation of this important cell type. Here we describe a two-step chemically defined xeno-free method for generating VEC-like cells from human pluripotent stem cells (hPSCs). HPSCs were specified to KDR
+ /ISL1+ multipotent cardiac progenitors (CPCs), followed by differentiation into valve endothelial-like cells (VELs) via an intermediate endocardial cushion cell (ECC) type. Mechanistically, administration of TGFb1 and BMP4 may specify VEC fate by activating the NOTCH/WNT signaling pathways and previously unidentified targets such as ATF3 and KLF family of transcription factors. When seeded onto the surface of the de-cellularized porcine aortic valve (DCV) matrix scaffolds, hPSC-derived VELs exhibit superior proliferative and clonogenic potential than the primary VECs and human aortic endothelial cells (HAEC). Our results show that hPSC-derived valvular cells could be efficiently generated from hPSCs, which might be used as seed cells for construction of valve organoids or next generation tissue engineered heart valves., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
7. Vitrification of Heart Valve Tissues.
- Author
-
Brockbank KGM, Chen Z, Greene ED, and Campbell LH
- Subjects
- Animals, Cell Survival, Heart Valves chemistry, Heart Valves drug effects, Humans, Phase Transition, Cryopreservation methods, Cryoprotective Agents pharmacology, Heart Valves cytology, Vitrification
- Abstract
Application of the original vitrification protocol used for pieces of heart valves to intact heart valves has evolved over time. Ice-free cryopreservation by Protocol 1 using VS55 is limited to small samples (1-3 mL total volume) where relatively rapid cooling and warming rates are possible. VS55 cryopreservation typically provides extracellular matrix preservation with approximately 80% cell viability and tissue function compared with fresh untreated tissues. In contrast, ice-free cryopreservation using VS83, Protocols 2 and 3, permits preservation of large samples (80-100 mL total volume) with several advantages over conventional cryopreservation methods and VS55 preservation, including long-term preservation capability at -80 °C; better matrix preservation than freezing with retention of material properties; very low cell viability, reducing the risks of an immune reaction in vivo; reduced risks of microbial contamination associated with use of liquid nitrogen; improved in vivo functions; no significant recipient allogeneic immune response; simplified manufacturing process; increased operator safety because liquid nitrogen is not used; and reduced manufacturing costs. More recently, we have developed Protocol 4 in which VS55 is supplemented with sugars resulting in reduced concerns regarding nucleation during cooling and warming. This method can be used for large samples resulting in retention of cell viability and permits short-term exposure to -80 °C with long-term storage preferred at or below -135 °C.
- Published
- 2021
- Full Text
- View/download PDF
8. Freeze-Drying of Decellularized Heart Valves for Off-the-Shelf Availability.
- Author
-
Wolkers WF and Hilfiker A
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Heart Valve Prosthesis, Heart Valves physiology, Swine, Cell Culture Techniques methods, Cryoprotective Agents chemistry, Extracellular Matrix chemistry, Freeze Drying methods, Heart Valves cytology, Tissue Engineering methods, Tissue Scaffolds chemistry
- Abstract
Malfunctioning heart valves can cause severe health problems, which if left untreated can lead to death. One of the treatment options is to replace a diseased heart valve with a decellularized valve construct prepared from human or animal material. Decellularized tissue scaffolds closely resemble properties of native tissue, while lacking immunogenic factors of cellular components. After transplantation, circulating stem and progenitor cells of the patient adhere to the scaffold resulting in in vivo tissue regeneration of the valve. Decellularized heart valve scaffold implants need to be stored to be readily available whenever needed, which can be done by freeze-drying. The advantage of freeze-drying is that it does not require bulky and energy-consuming freezing equipment for storage and allows easy transport. This chapter outlines the entire process from decellularization to freeze-drying to obtain dry decellularized heart valves, which after a simple rehydration step, can be used as implants. The protocol is described for porcine heart valves, but procedures can easily be adapted for material obtained from other species.
- Published
- 2021
- Full Text
- View/download PDF
9. Nfatc1 Promotes Interstitial Cell Formation During Cardiac Valve Development in Zebrafish.
- Author
-
Gunawan F, Gentile A, Gauvrit S, Stainier DYR, and Bensimon-Brito A
- Subjects
- Animals, Animals, Genetically Modified, Cell Movement physiology, Female, Male, Mice, Random Allocation, Zebrafish, Heart Valves cytology, Heart Valves growth & development, NFATC Transcription Factors physiology, Organogenesis physiology
- Abstract
Rationale: The transcription factor NFATC1 (nuclear factor of activated T-cell 1) has been implicated in cardiac valve formation in humans and mice, but we know little about the underlying mechanisms. To gain mechanistic understanding of cardiac valve formation at single-cell resolution and insights into the role of NFATC1 in this process, we used the zebrafish model as it offers unique attributes for live imaging and facile genetics., Objective: To understand the role of Nfatc1 in cardiac valve formation., Methods and Results: Using the zebrafish atrioventricular valve, we focus on the valve interstitial cells (VICs), which confer biomechanical strength to the cardiac valve leaflets. We find that initially atrioventricular endocardial cells migrate collectively into the cardiac jelly to form a bilayered structure; subsequently, the cells that led this migration invade the ECM (extracellular matrix) between the 2 endocardial cell monolayers, undergo endothelial-to-mesenchymal transition as marked by loss of intercellular adhesion, and differentiate into VICs. These cells proliferate and are joined by a few neural crest-derived cells. VIC expansion and a switch from a promigratory to an elastic ECM drive valve leaflet elongation. Functional analysis of Nfatc1 reveals its requirement during VIC development. Zebrafish nfatc1 mutants form significantly fewer VICs due to reduced proliferation and impaired recruitment of endocardial and neural crest cells during the early stages of VIC development. With high-speed microscopy and echocardiography, we show that reduced VIC formation correlates with valvular dysfunction and severe retrograde blood flow that persist into adulthood. Analysis of downstream effectors reveals that Nfatc1 promotes the expression of twist1b -a well-known regulator of epithelial-to-mesenchymal transition., Conclusions: Our study sheds light on the function of Nfatc1 in zebrafish cardiac valve development and reveals its role in VIC formation. It also further establishes the zebrafish as a powerful model to carry out longitudinal studies of valve formation and function.
- Published
- 2020
- Full Text
- View/download PDF
10. TGF-β Signaling Promotes Tissue Formation during Cardiac Valve Regeneration in Adult Zebrafish.
- Author
-
Bensimon-Brito A, Ramkumar S, Boezio GLM, Guenther S, Kuenne C, Helker CSM, Sánchez-Iranzo H, Iloska D, Piesker J, Pullamsetti S, Mercader N, Beis D, and Stainier DYR
- Subjects
- Animals, Cell Cycle, Endothelium metabolism, Extracellular Matrix metabolism, Heart Valves metabolism, Kidney metabolism, Models, Animal, Tissue Engineering methods, Zebrafish metabolism, Cell Differentiation, Endothelium cytology, Heart Valves cytology, Kidney cytology, Regeneration, Transforming Growth Factor beta metabolism, Zebrafish growth & development
- Abstract
Cardiac valve disease can lead to severe cardiac dysfunction and is thus a frequent cause of morbidity and mortality. Its main treatment is valve replacement, which is currently greatly limited by the poor recellularization and tissue formation potential of the implanted valves. As we still lack suitable animal models to identify modulators of these processes, here we used adult zebrafish and found that, upon valve decellularization, they initiate a rapid regenerative program that leads to the formation of new functional valves. After injury, endothelial and kidney marrow-derived cells undergo cell cycle re-entry and differentiate into new extracellular matrix-secreting valve cells. The TGF-β signaling pathway promotes the regenerative process by enhancing progenitor cell proliferation as well as valve cell differentiation. These findings reveal a key role for TGF-β signaling in cardiac valve regeneration and establish the zebrafish as a model to identify and test factors promoting cardiac valve recellularization and growth., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. In vivo tissue engineering of a trilayered leaflet-shaped tissue construct.
- Author
-
Jana S and Lerman A
- Subjects
- Animals, Collagen chemistry, Glycosaminoglycans chemistry, Polymers chemistry, Rats, Rats, Sprague-Dawley, Heart Valve Diseases therapy, Heart Valve Prosthesis, Heart Valve Prosthesis Implantation methods, Heart Valves cytology, Tissue Engineering methods, Tissue Scaffolds chemistry
- Abstract
Aim: We aimed to develop a leaflet-shaped trilayered tissue construct mimicking the morphology of native heart valve leaflets. Materials & methods: Electrospinning and in vivo tissue engineering methods were employed. Results: We developed leaflet-shaped microfibrous scaffolds, each with circumferentially, randomly and radially oriented three layers mimicking the trilayered, oriented structure of native leaflets. After 3 months in vivo tissue engineering with the scaffolds, the generated leaflet-shaped tissue constructs had a trilayered structure mimicking the orientations of native heart valve leaflets. Presence of collagen, glycosaminoglycans and elastin seen in native leaflets was observed in the engineered tissue constructs. Conclusion: Trilayered, oriented fibrous scaffolds brought the orientations of the infiltrated cells and their produced extracellular matrix proteins into the constructs.
- Published
- 2020
- Full Text
- View/download PDF
12. Dynamic Bioreactors with Integrated Microfabricated Devices for Mechanobiological Screening.
- Author
-
Beca BM, Sun Y, Wong E, Moraes C, and Simmons CA
- Subjects
- Actins metabolism, Animals, Biomechanical Phenomena, Heart Valves cytology, Hydrogels chemistry, Swine, Biophysics instrumentation, Bioreactors, Microtechnology instrumentation
- Abstract
Biomechanical stimulation is a common strategy to improve the growth, maturation, and function of a variety of engineered tissues. However, identifying optimized biomechanical conditioning protocols is challenging, as cell responses to mechanical stimuli are modulated by other multifactorial microenvironmental cues, including soluble factors and biomaterial properties. Traditional bioreactors lack the throughput necessary for combinatorial testing of cell activity in mechanically stimulated engineered tissues. Microfabricated systems can improve experimental throughput, but often do not provide uniform mechanical loading, are challenging to use, lack robustness, and offer limited amounts of cells and tissue for analysis. To address the need for higher-throughput, combinatorial testing of cell activity in a tissue engineering context, we developed a hybrid approach, in which flexible polydimethylsiloxane microfabricated inserts were designed to simultaneously generate multiple tensile strains when stretched cyclically in a standard dynamic bioreactor. In the embodiment presented in this study, each insert contained an array of 35 dog bone-shaped wells in which cell-seeded microscale hydrogels can be polymerized, with up to eight inserts stretched simultaneously in the bioreactor. Uniformity of the applied strains, both along the length of a microtissue and across multiple microtissues at the same strain level, was confirmed experimentally. In proof-of-principle experiments, the combinatorial effects of dynamic strain, biomaterial stiffness, and transforming growth factor (TGF)-β1 stimulation on myofibroblast differentiation were tested, revealing both known and novel interaction effects and suggesting tissue engineering strategies to regulate myofibroblast activation. This platform is expected to have wide applicability in systematically probing combinations of mechanobiological tissue engineering parameters for desired effects on cell fate and tissue function. Impact Statement In this study, we introduce a dynamic bioreactor system incorporating microfabricated inserts to enable systematic probing of the effects of combinations of mechanobiological parameters on engineered tissues. This novel platform offers the ease of use, robustness, and well-defined mechanical strain stimuli inherent in traditional dynamic bioreactors, but significantly improves throughput (up to 280 microtissues can be tested simultaneously in the embodiment presented in this study). This platform has wide applicability to systematically probe combinations of dynamic mechanical strain, biomaterial properties, biochemical stimulation, and other parameters for desired effects on cell fate and engineered tissue development.
- Published
- 2019
- Full Text
- View/download PDF
13. Optimizing detergent concentration and processing time to balance the decellularization efficiency and properties of bioprosthetic heart valves.
- Author
-
Luo Y, Lou D, Ma L, and Gao C
- Subjects
- Animals, Antigens metabolism, Cell Death drug effects, Cell Survival drug effects, DNA metabolism, Epitopes metabolism, Galactose metabolism, Mice, NIH 3T3 Cells, Swine, Bioprosthesis, Detergents pharmacology, Heart Valve Prosthesis, Heart Valves cytology
- Abstract
Decellularization treatment has been widely used to decrease the potential immunogenicity and improve the anticalcification properties of bio-derived materials, which may be utilized as an alternative method for the preparation of bioprosthetic heart valves. However, the excessive decellularization treatments will deteriarate the properties of heart valves. Among the decellularizaton parameters, detergent concentration and processing time are considered as those of the most key factors. Therefore, it should be meaningful to balance the decellularization efficiency and properties of bioprosthetic heart valves by optimizing the detergent concentration and processing time. In this study, three groups of the decellularized heart valves treated by sodium deoxycholate (SD) with different concentration and processing time were investigated through histological, biochemical, and mechanical analysis. Similar decellularization efficiency can be concluded through histological staining, DNA and α-Gal quantification results. Extracellular matrix contents quantification and tensile test results revealed that there is no obvious difference among the three decellularized heart valves. in vitro cytotoxicity assay showed that the remnant detergent is not enough to cause cell death, which indicated that the decellularized porcine aortic heart valves may be suitable for further in vivo research. In conclusion, Triton X-100/SD may be a suitable protocol used for heart valves decellularization. And it is feasible to vary the detergent processing time by changing the detergent concentration without compromising the decellularization efficiency., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
14. Quantifying heart valve interstitial cell contractile state using highly tunable poly(ethylene glycol) hydrogels.
- Author
-
Khang A, Gonzalez Rodriguez A, Schroeder ME, Sansom J, Lejeune E, Anseth KS, and Sacks MS
- Subjects
- Animals, Cells, Cultured, Heart Valves cytology, Interstitial Cells of Cajal cytology, Swine, Extracellular Matrix chemistry, Heart Valves metabolism, Hydrogels chemistry, Interstitial Cells of Cajal metabolism, Muscle Contraction, Polyethylene Glycols chemistry
- Abstract
Valve interstitial cells (VIC) are the primary cell type residing within heart valve tissues. In many valve pathologies, VICs become activated and will subsequently profoundly remodel the valve tissue extracellular matrix (ECM). A primary indicator of VIC activation is the upregulation of α-smooth muscle actin (αSMA) stress fibers, which in turn increase VIC contractility. Thus, contractile state reflects VIC activation and ECM biosynthesis levels. In general, cell contraction studies have largely utilized two-dimensional substrates, which are a vastly different micro mechanical environment than 3D native leaflet tissue. To address this limitation, hydrogels have been a popular choice for studying cells in a three-dimensional environment due to their tunable properties and optical transparency, which allows for direct cell visualization. In the present study, we extended the use of hydrogels to study the active contractile behavior of VICs. Aortic VICs (AVIC) were encapsulated within poly(ethylene glycol) (PEG) hydrogels and were subjected to flexural-deformation tests to assess the state of AVIC contraction. Using a finite element model of the experimental setup, we determined the effective shear modulus μ of the constructs. An increase in μ resulting from AVIC active contraction was observed. Results further indicated that AVIC contraction had a more pronounced effect on μ in softer gels (72 ± 21% increase in μ within 2.5 kPa gels) and was dependent upon the availability of adhesion sites (0.5-1 mM CRGDS). The transparency of the gel allowed us to image AVICs directly within the hydrogel, where we observed a time-dependent decrease in volume (time constant τ=3.04 min) when the AVICs were induced into a hypertensive state. Our results indicated that AVIC contraction was regulated by both the intrinsic (unseeded) gel stiffness and the CRGDS peptide concentrations. This finding suggests that AVIC contractile state can be profoundly modulated through their local micro environment using modifiable PEG gels in a 3D micromechanical-emulating environment. Moving forward, this approach has the potential to be used towards delineating normal and diseased VIC biomechanical properties using highly tunable PEG biomaterials. STATEMENT OF SIGNIFICANCE., (Copyright © 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
15. [The number and subgroups of lymphocytes in valve tissue of rheumatic heart disease combined with diabetes mellitus].
- Author
-
Wang H, Su J, Li J, Wang G, and Xiu L
- Subjects
- Case-Control Studies, Humans, B-Lymphocytes cytology, CD8-Positive T-Lymphocytes cytology, Diabetes Mellitus pathology, Heart Valves cytology, Rheumatic Heart Disease complications, T-Lymphocytes, Regulatory cytology
- Abstract
Objective To investigate the effect of diabetes mellitus on lymphocytes in rheumatic heart valve tissue and its mechanism. Methods Valve tissues of 40 patients undergoing heart valve replacement were collected, including 20 patients in rheumatic heart disease group (without diabetes) and 20 patients in diabetic group (rheumatic heart disease combined with diabetes). In addition, 20 cases of valve tissue from control group were collected. HE staining was used to observe the damage of valve tissue and the area of collagen degeneration. CD4
+ T cells, CD8+ T cells, B cells and plasma cells were detected by immunohistochemical staining. Flow cytometry was used to detect the proportion of regulatory T cells (Tregs) in peripheral blood. Results Compared with the rheumatic heart disease group, the damage of valve tissue in the diabetic group was further aggravated, the number of infiltrating inflammatory cells increased, and the area of collagen degeneration was enlarged. Compared with the control group, the number of T cells, CD4+ T cells, CD8+ T cells, B cells and plasma cells in valve tissue of patients with rheumatic heart disease increased significantly. Diabetes mellitus further increased the number of T cells, CD4+ T cells, B cells and plasma cells in valve tissue, but had no significant effect on CD8+ T cells. The proportion of Tregs in the peripheral blood of patients with rheumatic heart disease was significantly reduced. Diabetes mellitus could further reduce the proportion of Tregs. Conclusion The number of T cells, CD4+ T cells, B cells and plasma cells in heart valves of rheumatic heart disease patients with diabetes mellitus go up significantly, and Treg ratio goes down.- Published
- 2019
16. Biologically Inspired Scaffolds for Heart Valve Tissue Engineering via Melt Electrowriting.
- Author
-
Saidy NT, Wolf F, Bas O, Keijdener H, Hutmacher DW, Mela P, and De-Juan-Pardo EM
- Subjects
- Biomechanical Phenomena, Biomimetics methods, Blood Vessel Prosthesis, Cells, Cultured, Guided Tissue Regeneration instrumentation, Guided Tissue Regeneration methods, Heart Valve Diseases pathology, Heart Valve Diseases therapy, Humans, Infant, Newborn, Materials Testing, Myocytes, Smooth Muscle cytology, Polymers chemistry, Umbilical Cord cytology, Biomimetics instrumentation, Electroplating methods, Heart Valves cytology, Printing, Three-Dimensional, Tissue Engineering instrumentation, Tissue Engineering methods, Tissue Scaffolds chemistry
- Abstract
Heart valves are characterized to be highly flexible yet tough, and exhibit complex deformation characteristics such as nonlinearity, anisotropy, and viscoelasticity, which are, at best, only partially recapitulated in scaffolds for heart valve tissue engineering (HVTE). These biomechanical features are dictated by the structural properties and microarchitecture of the major tissue constituents, in particular collagen fibers. In this study, the unique capabilities of melt electrowriting (MEW) are exploited to create functional scaffolds with highly controlled fibrous microarchitectures mimicking the wavy nature of the collagen fibers and their load-dependent recruitment. Scaffolds with precisely-defined serpentine architectures reproduce the J-shaped strain stiffening, anisotropic and viscoelastic behavior of native heart valve leaflets, as demonstrated by quasistatic and dynamic mechanical characterization. They also support the growth of human vascular smooth muscle cells seeded both directly or encapsulated in fibrin, and promote the deposition of valvular extracellular matrix components. Finally, proof-of-principle MEW trileaflet valves display excellent acute hydrodynamic performance under aortic physiological conditions in a custom-made flow loop. The convergence of MEW and a biomimetic design approach enables a new paradigm for the manufacturing of scaffolds with highly controlled microarchitectures, biocompatibility, and stringent nonlinear and anisotropic mechanical properties required for HVTE., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
17. Impact of modified gelatin on valvular microtissues.
- Author
-
Roosens A, Handoyo YP, Dubruel P, and Declercq H
- Subjects
- Animals, Extracellular Matrix Proteins biosynthesis, Heart Valves cytology, Swine, Cell Movement drug effects, Cell Proliferation drug effects, Gelatin chemistry, Gelatin pharmacology, Gene Expression Regulation drug effects, Heart Valves metabolism, Hydrogels chemistry, Hydrogels pharmacology
- Abstract
A significant challenge in the field of tissue engineering is the biofabrication of three-dimensional (3D) functional tissues with direct applications in organ-on-a-chip systems and future organ engineering. Multicellular valvular microtissues can be used as building blocks for the formation of larger scale valvular macrotissues. Yet, for the controlled biofabrication of 3D macrotissues with predefined complex shapes, directed assembly of microtissues through bioprinting is needed. This study aimed to investigate if modified gelatin is an instructive material for valvular microtissues. Valvular microtissues were encapsulated in modified gelatin hydrogels and cross-linked in the presence of a photoinitiator (Irgacure 2959 or VA-086). Hydrogel properties were determined, and valvular interstitial cell functions like phenotype, proliferation, migration, mRNA expression of extracellular matrix (ECM) molecules, ECM deposition, and tissue fusion were characterized by histochemical stainings and RT-qPCR. Encapsulated microtissues remained viable, produced heart valve-related ECM components, and remained in a quiescent state. However, encapsulation induced some changes in ECM formation and gene expression. Encapsulated microtissues showed lower remodeling capacity and increased expression levels of Col I/V, elastin, hyaluronan, biglycan, decorin, and Sox9 compared with nonencapsulated microtissues. Furthermore, this study demonstrated that proliferation, migration, and tissue fusion was more pronounced in softer gels. In general, we evidenced that modified gelatin is an instructive material for physiologically relevant valvular microtissues and provided a proof of concept for the formation of larger valvular tissue by assembling microtissues at random in soft gels., (© 2019 John Wiley & Sons, Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
18. Non-pathological Chondrogenic Features of Valve Interstitial Cells in Normal Adult Zebrafish.
- Author
-
Schulz A, Brendler J, Blaschuk O, Landgraf K, Krueger M, and Ricken AM
- Subjects
- Animals, Cartilage cytology, Collagen analysis, Heart Valves growth & development, Heart Valves pathology, Mice, Mice, Inbred C57BL, Species Specificity, Zebrafish, Aging, Chondrogenesis, Heart Valves cytology, Heart Valves ultrastructure
- Abstract
In the heart, unidirectional blood flow depends on proper heart valve function. As, in mammals, regulatory mechanisms of early heart valve and bone development are shown to contribute to adult heart valve pathologies, we used the animal model zebrafish (ZF, Danio rerio) to investigate the microarchitecture and differentiation of cardiac valve interstitial cells in the transition from juvenile (35 days) to end of adult breeding (2.5 years) stages. Of note, light microscopy and immunohistochemistry revealed major differences in ZF heart valve microarchitecture when compared with adult mice. We demonstrate evidence for rather chondrogenic features of valvular interstitial cells by histological staining and immunodetection of SOX-9, aggrecan, and type 2a1 collagen. Collagen depositions are enriched in a thin layer at the atrial aspect of atrioventricular valves and the ventricular aspect of bulboventricular valves, respectively. At the ultrastructural level, the collagen fibrils are lacking obvious periodicity and orientation throughout the entire valve.
- Published
- 2019
- Full Text
- View/download PDF
19. Endothelial-to-Mesenchymal Transition.
- Author
-
Bischoff J
- Subjects
- Age Factors, Animals, Cardiovascular Diseases etiology, Cell Movement physiology, Endothelial Cells drug effects, Endothelial Cells physiology, Epithelial-Mesenchymal Transition drug effects, Extracellular Matrix metabolism, Heart Valves cytology, Heart Valves growth & development, Leukocyte Common Antigens metabolism, Mice, Mitral Valve, Mitral Valve Insufficiency etiology, Models, Animal, Myocardial Infarction complications, Neovascularization, Physiologic, Sheep, Stress, Physiological, Transforming Growth Factor beta pharmacology, Epithelial-Mesenchymal Transition physiology
- Published
- 2019
- Full Text
- View/download PDF
20. Endocardially Derived Macrophages Are Essential for Valvular Remodeling.
- Author
-
Shigeta A, Huang V, Zuo J, Besada R, Nakashima Y, Lu Y, Ding Y, Pellegrini M, Kulkarni RP, Hsiai T, Deb A, Zhou B, Nakano H, and Nakano A
- Subjects
- Animals, Embryo, Mammalian metabolism, Hematopoiesis physiology, Mesoderm metabolism, Mice, Transgenic, NFATC Transcription Factors metabolism, Yolk Sac, Endocardium metabolism, Gene Expression Regulation, Developmental physiology, Heart Valves cytology, Macrophages metabolism
- Abstract
During mammalian embryogenesis, de novo hematopoiesis occurs transiently in multiple anatomical sites including the yolk sac, dorsal aorta, and heart tube. A long-unanswered question is whether these local transient hematopoietic mechanisms are essential for embryonic growth. Here, we show that endocardial hematopoiesis is critical for cardiac valve remodeling as a source of tissue macrophages. Colony formation assay from explanted heart tubes and genetic lineage tracing with the endocardial specific Nfatc1-Cre mouse revealed that hemogenic endocardium is a de novo source of tissue macrophages in the endocardial cushion, the primordium of the cardiac valves. Surface marker characterization, gene expression profiling, and ex vivo phagocytosis assay revealed that the endocardially derived cardiac tissue macrophages play a phagocytic and antigen presenting role. Indeed, genetic ablation of endocardially derived macrophages caused severe valve malformation. Together, these data suggest that transient hemogenic activity in the endocardium is indispensable for the valvular tissue remodeling in the heart., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. VGLL4 plays a critical role in heart valve development and homeostasis.
- Author
-
Yu W, Ma X, Xu J, Heumüller AW, Fei Z, Feng X, Wang X, Liu K, Li J, Cui G, Peng G, Ji H, Li J, Jing N, Song H, Lin Z, Zhao Y, Wang Z, Zhou B, and Zhang L
- Subjects
- Animals, Cell Lineage, Cell Proliferation, Endothelial Cells metabolism, Epithelial-Mesenchymal Transition, Gene Expression Regulation, Developmental, Gene Knockout Techniques, Heart Valves cytology, Heart Valves metabolism, Hippo Signaling Pathway, Homeostasis, Hypertrophy, Left Ventricular veterinary, Mice, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Endothelial Cells cytology, Heart Valves growth & development, Hypertrophy, Left Ventricular genetics, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Heart valve disease is a major clinical problem worldwide. Cardiac valve development and homeostasis need to be precisely controlled. Hippo signaling is essential for organ development and tissue homeostasis, while its role in valve formation and morphology maintenance remains unknown. VGLL4 is a transcription cofactor in vertebrates and we found it was mainly expressed in valve interstitial cells at the post-EMT stage and was maintained till the adult stage. Tissue specific knockout of VGLL4 in different cell lineages revealed that only loss of VGLL4 in endothelial cell lineage led to valve malformation with expanded expression of YAP targets. We further semi-knockout YAP in VGLL4 ablated hearts, and found hyper proliferation of arterial valve interstitial cells was significantly constrained. These findings suggest that VGLL4 is important for valve development and manipulation of Hippo components would be a potential therapy for preventing the progression of congenital valve disease., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
22. Behavior of valvular interstitial cells on trilayered nanofibrous substrate mimicking morphologies of heart valve leaflet.
- Author
-
Jana S and Lerman A
- Subjects
- Animals, Cell Proliferation, Cell Shape, Cell Survival, Collagen chemistry, Gene Expression Regulation, Heart Valves ultrastructure, Humans, Nanofibers toxicity, Nanofibers ultrastructure, Sus scrofa, Tensile Strength, Tissue Scaffolds chemistry, Heart Valves anatomy & histology, Heart Valves cytology, Nanofibers chemistry
- Abstract
Heart valve tissue engineering could be an alternative to the current bioprosthetic heart valve that faces limitations especially in pediatric patients. However, heart valve tissue engineering has remained challenging because leaflets - the primary component of a heart valve - have three layers with three diverse orientations - circumferential, random and radial, respectively. In order to mimic the orientations, we first designed three novel collectors to fabricate three nanofibrous layers with those orientations from a polymeric biomaterial in an electrospinning system. Then, we devised a novel direct electrospinning technique to develop a unified trilayered nanofibrous (TN) substrate comprising those oriented layers. The TN substrate supported the growth and orientations of seeded porcine valvular interstitial cells (PVICs) and their deposited collagen fibrils. After one month culture, the obtained trilayered tissue construct (TC) exhibited increased tensile properties over its TN substrate. Most importantly, the developed TC did not show any sign of shrinkage. Gene expression pattern of the PVICs indicated the developing stage of the TC. Their protein expression pattern was quite similar to that of leaflets. STATEMENT OF SIGNIFICANCE: This manuscript talks about development of a novel trilayered nanofibrous substrate mimicking the morphologies of a heart valve leaflet. It also describes culturing of valvular interstitial cells that reside in a leaflet, in the substrate and compares the behavior of the cultured cells with that in native leaflets in terms cell morphology, protein deposition and its orientation, and molecular signature. This study builds the groundwork for our future trilayered, tissue-engineered leaflet development. This research article would be of great interest to investigators and researchers in the field of cardiovascular tissue engineering especially in cardiac valve tissue engineering through biomaterial-based tissue engineering., (Copyright © 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
23. Dynamic Expression Profiles of Sox9 in Embryonic, Post Natal, and Adult Heart Valve Cell Populations.
- Author
-
Gallina D and Lincoln J
- Subjects
- Animals, Animals, Newborn, Embryonic Stem Cells cytology, Endothelium, Vascular cytology, Gene Expression Regulation, Developmental, Heart Valves cytology, Mesenchymal Stem Cells cytology, Mice, Mice, Inbred C57BL, SOX9 Transcription Factor genetics, Embryonic Stem Cells metabolism, Endothelium, Vascular metabolism, Heart Valves metabolism, Mesenchymal Stem Cells metabolism, SOX9 Transcription Factor metabolism
- Abstract
Heart valves are dynamic structures and abnormalities during embryonic development can lead to premature lethality or congenital malformations present at birth. The transcription factor Sox9 has been shown to be critical for early and late stages of valve formation, but its defined expression pattern throughout embryonic, post natal, and adult growth and maturation is incomplete. Here we use an antibody to detect 1-100 amino acids of Sox9 and show that in the developing embryo, Sox9 is not detected in valve endothelial cells (VECs) lining the primitive valve structures, but is highly expressed in the endothelial-derived valve interstitial cell population following endothelial-to-mesenchymal transformation. Expression is maintained in this cell population after birth, but is additionally detected in VECs from post natal day 1. Using a specific antibody to detect a phosphorylated form of Sox9 at Serine 181 (pSox9), we note enrichment of pSox9 in VECs at post natal days 1 and 10 and this pattern correlates with the known upstream kinase RockI, and downstream target, Aggrecan. The contribution of Sox9 to post natal growth and maturation of the valve is not known, but this study provides insights for future work examining the differential functions of Sox9 protein in valve cell populations. Anat Rec, 302:108-116, 2019. © 2018 Wiley Periodicals, Inc., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
24. Correlation between valvular interstitial cell morphology and phenotypes: A novel way to detect activation.
- Author
-
Ali MS, Deb N, Wang X, Rahman M, Christopher GF, and Lacerda CMR
- Subjects
- Animals, Heart Valves metabolism, Myofibroblasts cytology, Myofibroblasts metabolism, Phenotype, Swine, Cell Differentiation physiology, Fibroblasts cytology, Fibroblasts metabolism, Heart Valves cytology
- Abstract
Valvular interstitial cells (VICs) constitute the major cell population in heart valves. Quiescent fibroblastic VICs are seen in adult healthy valves. They become activated myofibroblastic VICs during development, in diseased valves and in vitro. 2D substrate stiffness within a 5-15 kPa range along with high passage numbers promote VIC activation in vitro. In this study, we characterize VIC quiescence and activation across a 1-21 kPa range of substrate stiffness and passages. We define a cell morphology characterization system for VICs as they transform. We hypothesize that VICs show distinct morphological characteristics in different activation states and the morphology distribution varies with substrate stiffness and passage number. Four VIC morphologies - tailed, spindle, rhomboid and triangle - account for the majority of VIC in this study. Using α-smooth muscle actin (α-SMA), non-muscle myosin heavy chain B (SMemb) and transforming growth factor β (TGF-β) as activation markers for validation, we developed a system where we categorize morphology distribution of VIC cultures, to be potentially used as a non-destructive detection method of activation state. We also show that this system can be used to force stiffness-induced deactivation. The reversibility in VIC activation has important implications in in vitro research and tissue engineering., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
25. Bmp2 and Notch cooperate to pattern the embryonic endocardium.
- Author
-
Papoutsi T, Luna-Zurita L, Prados B, Zaffran S, and de la Pompa JL
- Subjects
- Animals, Bone Morphogenetic Protein 2 genetics, Embryo, Mammalian cytology, Endocardium cytology, Heart Valves cytology, Mice, Mice, Transgenic, Myocardium cytology, Myocardium metabolism, Receptors, Notch genetics, Smad1 Protein genetics, Smad1 Protein metabolism, Smad5 Protein genetics, Smad5 Protein metabolism, Bone Morphogenetic Protein 2 metabolism, Embryo, Mammalian embryology, Endocardium embryology, Heart Valves embryology, Receptors, Notch metabolism, Signal Transduction physiology
- Abstract
Signaling interactions between the myocardium and endocardium pattern embryonic cardiac regions, instructing their development to fulfill specific functions in the mature heart. We show that ectopic Bmp2 expression in the mouse chamber myocardium changes the transcriptional signature of adjacent chamber endocardial cells into valve tissue, and enables them to undergo epithelial-mesenchyme transition. This induction is independent of valve myocardium specification and requires high levels of Notch1 activity. Biochemical experiments suggest that Bmp2-mediated Notch1 induction is achieved through transcriptional activation of the Notch ligand Jag1, and physical interaction of Smad1/5 with the intracellular domain of the Notch1 receptor. Thus, widespread myocardial Bmp2 and endocardial Notch signaling drive presumptive ventricular endocardium to differentiate into valve endocardium. Understanding the molecular basis of valve development is instrumental to designing therapeutic strategies for congenital heart valve defects., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
26. Fabrication and In Vitro Characterization of a Tissue Engineered PCL-PLLA Heart Valve.
- Author
-
Hasan A, Soliman S, El Hajj F, Tseng YT, Yalcin HC, and Marei HE
- Subjects
- Animals, Cell Survival, Cells, Cultured, Equipment Design, Female, Humans, Mice, Inbred C57BL, Swine, Tissue Engineering methods, Bioprosthesis, Heart Valve Prosthesis, Heart Valves cytology, Polyesters chemistry, Stem Cells cytology, Tissue Scaffolds chemistry
- Abstract
Heart valve diseases are among the leading causes of cardiac failure around the globe. Nearly 90,000 heart valve replacements occur in the USA annually. Currently, available options for heart valve replacement include bioprosthetic and mechanical valves, both of which have severe limitations. Bioprosthetic valves can last for only 10-20 years while patients with mechanical valves always require blood-thinning medications throughout the remainder of the patient's life. Tissue engineering has emerged as a promising solution for the development of a viable, biocompatible and durable heart valve; however, a human implantable tissue engineered heart valve is yet to be achieved. In this study, a tri-leaflet heart valve structure is developed using electrospun polycaprolactone (PCL) and poly L-lactic acid (PLLA) scaffolds, and a set of in vitro testing protocol has been developed for routine manufacturing of tissue engineered heart valves. Stress-strain curves were obtained for mechanical characterization of different valves. The performances of the developed valves were hemodynamically tested using a pulse duplicator, and an echocardiography machine. Results confirmed the superiority of the PCL-PLLA heart valve compared to pure PCL or pure PLLA. The developed in vitro test protocol involving pulse duplicator and echocardiography tests have enormous potential for routine application in tissue engineering of heart valves.
- Published
- 2018
- Full Text
- View/download PDF
27. Human iPSC-derived mesenchymal stem cells encapsulated in PEGDA hydrogels mature into valve interstitial-like cells.
- Author
-
Nachlas ALY, Li S, Jha R, Singh M, Xu C, and Davis ME
- Subjects
- Antigens, Differentiation biosynthesis, Cells, Immobilized cytology, Heart Valves cytology, Humans, Induced Pluripotent Stem Cells cytology, Mesenchymal Stem Cells cytology, Cell Differentiation, Cells, Immobilized metabolism, Heart Valves metabolism, Induced Pluripotent Stem Cells metabolism, Mesenchymal Stem Cells metabolism, Polyethylene Glycols chemistry
- Abstract
Despite recent advances in tissue engineered heart valves (TEHV), a major challenge is identifying a cell source for seeding TEHV scaffolds. Native heart valves are durable because valve interstitial cells (VICs) maintain tissue homeostasis by synthesizing and remodeling the extracellular matrix. This study demonstrates that induced pluripotent stem cells (iPSC)-derived mesenchymal stem cells (iMSCs) can be derived from iPSCs using a feeder-free protocol and then further matured into VICs by encapsulation within 3D hydrogels. The differentiation efficiency was characterized using flow cytometry, immunohistochemistry staining, and trilineage differentiation. Using our feeder-free differentiation protocol, iMSCs were differentiated from iPSCs and had CD90
+ , CD44+ , CD71+ , αSMA+ , and CD45- expression. Furthermore, iMSCs underwent trilineage differentiation when cultured in induction media for 21 days. iMSCs were then encapsulated in poly(ethylene glycol)diacrylate (PEGDA) hydrogels grafted with adhesion peptide (RGDS) to promote remodeling and further maturation into VIC-like cells. VIC phenotype was assessed by the expression of alpha-smooth muscle actin (αSMA), vimentin, and collagen production after 28 days. When MSC-derived cells were encapsulated in PEGDA hydrogels that mimic the leaflet modulus, a decrease in αSMA expression and increase in vimentin was observed. In addition, iMSCs synthesized collagen type I after 28 days in 3D hydrogel culture. Thus, the results from this study suggest that iMSCs may be a promising cell source for TEHV., Statement of Significance: Developing a suitable cell source is a critical component for the success and durability of tissue engineered heart valves. The significance of this study is the generation of iPSCs-derived mesenchymal stem cells (iMSCs) that have the capacity to mature into valve interstitial-like cells when introduced into a 3D cell culture designed to mimic the layers of the valve leaflet. iMSCs were generated using a feeder-free protocol, which is one major advantage over other methods, as it is more clinically relevant. In addition to generating a potential new cell source for heart valve tissue engineering, this study also highlights the importance of a 3D culture environment to influence cell phenotype and function., (Copyright © 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
28. A human pericardium biopolymeric scaffold for autologous heart valve tissue engineering: cellular and extracellular matrix structure and biomechanical properties in comparison with a normal aortic heart valve.
- Author
-
Straka F, Schornik D, Masin J, Filova E, Mirejovsky T, Burdikova Z, Svindrych Z, Chlup H, Horny L, Daniel M, Machac J, Skibová J, Pirk J, and Bacakova L
- Subjects
- Biomechanical Phenomena, Biopolymers chemistry, Humans, Materials Testing, Tensile Strength, Aorta, Extracellular Matrix metabolism, Heart Valves cytology, Mechanical Phenomena, Pericardium cytology, Tissue Engineering, Tissue Scaffolds chemistry
- Abstract
The objective of our study was to compare the cellular and extracellular matrix (ECM) structure and the biomechanical properties of human pericardium (HP) with the normal human aortic heart valve (NAV). HP tissues (from 12 patients) and NAV samples (from 5 patients) were harvested during heart surgery. The main cells in HP were pericardial interstitial cells, which are fibroblast-like cells of mesenchymal origin similar to the valvular interstitial cells in NAV tissue. The ECM of HP had a statistically significantly (p < 0.001) higher collagen I content, a lower collagen III and elastin content, and a similar glycosaminoglycans (GAGs) content, in comparison with the NAV, as measured by ECM integrated density. However, the relative thickness of the main load-bearing structures of the two tissues, the dense part of fibrous HP (49 ± 2%) and the lamina fibrosa of NAV (47 ± 4%), was similar. In both tissues, the secant elastic modulus (Es) was significantly lower in the transversal direction (p < 0.05) than in the longitudinal direction. This proved that both tissues were anisotropic. No statistically significant differences in UTS (ultimate tensile strength) values and in calculated bending stiffness values in the longitudinal or transversal direction were found between HP and NAV. Our study confirms that HP has an advantageous ECM biopolymeric structure and has the biomechanical properties required for a tissue from which an autologous heart valve replacement may be constructed.
- Published
- 2018
- Full Text
- View/download PDF
29. MicroRNA-449c-5p inhibits osteogenic differentiation of human VICs through Smad4-mediated pathway.
- Author
-
Xu R, Zhao M, Yang Y, Huang Z, Shi C, Hou X, Zhao Y, Chen B, Xiao Z, Liu J, Miao Q, and Dai J
- Subjects
- 3' Untranslated Regions, Animals, Aortic Valve metabolism, Aortic Valve pathology, Aortic Valve Stenosis genetics, Aortic Valve Stenosis metabolism, Aortic Valve Stenosis pathology, Biomarkers, Calcinosis genetics, Calcinosis metabolism, Calcinosis pathology, Cells, Cultured, Connective Tissue Cells cytology, Female, Gene Expression Profiling, Gene Expression Regulation, Gene Knockdown Techniques, Humans, Male, Mice, Phenotype, RNA Interference, Smad4 Protein genetics, Cell Differentiation genetics, Connective Tissue Cells metabolism, Heart Valves cytology, MicroRNAs genetics, Osteogenesis genetics, Signal Transduction, Smad4 Protein metabolism
- Abstract
Calcific aortic valve disease (CAVD) is the most common heart valve disorder, yet its mechanism remains poorly understood. Valve interstitial cells (VICs) are the prevalent cells in aortic valve and their osteogenic differentiation may be responsible for calcific nodule formation in CAVD pathogenesis. Emerging evidence shows microRNA (miRNA, or miR) can function as important regulators of many pathological processes, including osteogenic differentiation. Here, we aimed to explore the function of miR-449c-5p in CAVD pathogenesis. In this study, we demonstrated the role of miR-449c-5p in VICs osteogenesis. MiRNA microarray assay and qRT-PCR results revealed miR-449c-5p was significantly down-regulated in calcified aortic valves compared with non-calcified valves. MiR-449c-5p overexpression inhibited VICs osteogenic differentiation in vitro, whereas down-regulation of miR-449c-5p enhanced the process. Target prediction analysis and dual-luciferase reporter assay confirmed Smad4 was a direct target of miR-449c-5p. Furthermore, knockdown of Smad4 inhibited VICs osteogenic differentiation, similar to the effect observed in up-regulation miR-449c-5p. In addition, animal experiments proved indirectly miR-449c-5p could alleviate aortic valve calcification. Our data suggested miR-449c-5p could function as a new inhibitory regulator of VICs osteogenic differentiation, which may act by targeting Smad4. MiR-449c-5p may be a potential therapeutic target for CAVD.
- Published
- 2017
- Full Text
- View/download PDF
30. Valve interstitial cell culture: Production of mature type I collagen and precise detection.
- Author
-
Liskova J, Hadraba D, Filova E, Konarik M, Pirk J, Jelen K, and Bacakova L
- Subjects
- Animals, Cell Culture Techniques, Cells, Cultured, Collagen Type I analysis, Collagen Type I genetics, Heart Valves chemistry, Heart Valves metabolism, Leydig Cells metabolism, Male, Staining and Labeling, Swine, Collagen Type I metabolism, Heart Valves cytology, Leydig Cells chemistry
- Abstract
Collagen often acts as an extracellular and intracellular marker for in vitro experiments, and its quality defines tissue constructs. To validate collagen detection techniques, cardiac valve interstitial cells were isolated from pigs and cultured under two different conditions; with and without ascorbic acid. The culture with ascorbic acid reached higher cell growth and collagen deposition, although the expression levels of collagen gene stayed similar to the culture without ascorbic acid. The fluorescent microscopy was positive for collagen fibers in both the cultures. Visualization of only extracellular collagen returned a higher correlation coefficient when comparing the immunolabeling and second harmonic generation microscopy images in the culture with ascorbic acid. Lastly, it was proved that the hydroxyproline strongly contributes to the second-order susceptibility tensor of collagen molecules, and therefore the second harmonic generation signal is impaired in the culture without ascorbic acid., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. Myofibroblastic activation of valvular interstitial cells is modulated by spatial variations in matrix elasticity and its organization.
- Author
-
Ma H, Killaars AR, DelRio FW, Yang C, and Anseth KS
- Subjects
- Actins analysis, Actins metabolism, Adaptor Proteins, Signal Transducing analysis, Adaptor Proteins, Signal Transducing metabolism, Animals, Biocompatible Materials metabolism, Biomechanical Phenomena, Cell Proliferation, Cells, Cultured, Elastic Modulus, Fibroblasts metabolism, Heart Valves metabolism, Hydrogels metabolism, Mechanotransduction, Cellular, Myofibroblasts metabolism, Polyethylene Glycols metabolism, Swine, Extracellular Matrix metabolism, Fibroblasts cytology, Heart Valves cytology, Myofibroblasts cytology
- Abstract
Valvular interstitial cells (VICs) are key regulators of the heart valve's extracellular matrix (ECM), and upon tissue damage, quiescent VIC fibroblasts become activated to myofibroblasts. As the behavior of VICs during disease progression and wound healing is different compared to healthy tissue, we hypothesized that the organization of the matrix mechanics, which results from depositing of collagen fibers, would affect VIC phenotypic transition. Specifically, we investigated how the subcellular organization of ECM mechanical properties affects subcellular localization of Yes-associated protein (YAP), an early marker of mechanotransduction, and α-smooth muscle actin (α-SMA), a myofibroblast marker, in VICs. Photo-tunable hydrogels were used to generate substrates with different moduli and to create organized and disorganized patterns of varying elastic moduli. When porcine VICs were cultured on these matrices, YAP and α-SMA activation were significantly increased on substrates with higher elastic modulus or a higher percentage of stiff regions. Moreover, VICs cultured on substrates with a spatially disorganized elasticity had smaller focal adhesions, less nuclear localized YAP, less α-SMA organization into stress fibers and higher proliferation compared to those cultured on substrates with a regular mechanical organization. Collectively, these results suggest that disorganized spatial variations in mechanics that appear during wound healing and fibrotic disease progression may influence the maintenance of the VIC fibroblast phenotype, causing more proliferation, ECM remodeling and matrix deposition., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Formation and function of intracardiac valve cells in the Drosophila heart.
- Author
-
Lammers K, Abeln B, Hüsken M, Lehmacher C, Psathaki OE, Alcorta E, Meyer H, and Paululat A
- Subjects
- Animals, Drosophila melanogaster cytology, Drosophila melanogaster growth & development, Heart Valves cytology, Heart Valves physiology, Heart Valves ultrastructure, Larva cytology, Larva physiology, Microscopy, Electron, Transmission, Myocytes, Cardiac physiology, Myocytes, Cardiac ultrastructure, Myofibrils, Drosophila melanogaster physiology, Myocytes, Cardiac cytology
- Abstract
Drosophila harbours a simple tubular heart that ensures haemolymph circulation within the body. The heart is built by a few different cell types, including cardiomyocytes that define the luminal heart channel and ostia cells that constitute openings in the heart wall allowing haemolymph to enter the heart chamber. Regulation of flow directionality within a tube, such as blood flow in arteries or insect haemolymph within the heart lumen, requires a dedicated gate, valve or flap-like structure that prevents backflow of fluids. In the Drosophila heart, intracardiac valves provide this directionality of haemolymph streaming, with one valve being present in larvae and three valves in the adult fly. Each valve is built by two specialised cardiomyocytes that exhibit a unique histology. We found that the capacity to open and close the heart lumen relies on a unique myofibrillar setting as well as on the presence of large membranous vesicles. These vesicles are of endocytic origin and probably represent unique organelles of valve cells. Moreover, we characterised the working mode of the cells in real time. Valve cells exhibit a highly flexible shape and, during each heartbeat, oscillating shape changes result in closing and opening of the heart channel. Finally, we identified a set of novel valve cell markers useful for future in-depth analyses of cell differentiation in wild-type and mutant animals., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
33. Biological and mechanical evaluation of a Bio-Hybrid scaffold for autologous valve tissue engineering.
- Author
-
Jahnavi S, Saravanan U, Arthi N, Bhuvaneshwar GS, Kumary TV, Rajan S, and Verma RS
- Subjects
- Animals, Biomechanical Phenomena, Cattle, Cell Communication, Cells, Cultured, Collagen metabolism, Fluorescent Antibody Technique, Heart Valves cytology, Humans, Microscopy, Electron, Scanning, Pericardium metabolism, Stress, Mechanical, Transplantation, Autologous, Heart Valve Prosthesis, Heart Valves physiology, Tissue Engineering methods, Tissue Scaffolds chemistry
- Abstract
Major challenge in heart valve tissue engineering for paediatric patients is the development of an autologous valve with regenerative capacity. Hybrid tissue engineering approach is recently gaining popularity to design scaffolds with desired biological and mechanical properties that can remodel post implantation. In this study, we fabricated aligned nanofibrous Bio-Hybrid scaffold made of decellularized bovine pericardium: polycaprolactone-chitosan with optimized polymer thickness to yield the desired biological and mechanical properties. CD44
+ , αSMA+ , Vimentin+ and CD105- human valve interstitial cells were isolated and seeded on these Bio-Hybrid scaffolds. Subsequent biological evaluation revealed interstitial cell proliferation with dense extra cellular matrix deposition that indicated the viability for growth and proliferation of seeded cells on the scaffolds. Uniaxial mechanical tests along axial direction showed that the Bio-Hybrid scaffolds has at least 20 times the strength of the native valves and its stiffness is nearly 3 times more than that of native valves. Biaxial and uniaxial mechanical studies on valve interstitial cells cultured Bio-Hybrid scaffolds revealed that the response along the axial and circumferential direction was different, similar to native valves. Overall, our findings suggest that Bio-Hybrid scaffold is a promising material for future development of regenerative heart valve constructs in children., (Copyright © 2016 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
34. A survey of membrane receptor regulation in valvular interstitial cells cultured under mechanical stresses.
- Author
-
Ali MS, Wang X, and Lacerda CM
- Subjects
- Animals, Biomechanical Phenomena, Core Binding Factor Alpha 1 Subunit genetics, Core Binding Factor Alpha 1 Subunit metabolism, Cytokines genetics, Cytokines metabolism, Gene Expression Profiling, Heart Valves cytology, Myofibroblasts cytology, Primary Cell Culture, Stress, Mechanical, Swine, Thrombospondin 1 genetics, Thrombospondin 1 metabolism, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Vascular Endothelial Growth Factor Receptor-3 genetics, Vascular Endothelial Growth Factor Receptor-3 metabolism, Gene Expression Regulation, Gene Regulatory Networks, Heart Valves metabolism, Mechanotransduction, Cellular genetics, Myofibroblasts metabolism
- Abstract
Degenerative valvular diseases have been linked to the action of abnormal forces on valve tissues during each cardiac cycle. It is now accepted that the degenerative behavior of valvular cells can be induced mechanically in vitro. This approach of in vitro modeling of valvular cells in culture constitutes a powerful tool to study, characterize, and develop predictors of heart valve degeneration in vivo. Using such in vitro systems, we expect to determine the exact signaling mechanisms that trigger and mediate propagation of degenerative signals. In this study, we aim to uncover the role of mechanosensing proteins on valvular cell membranes. These can be cell receptors and triggers of downstream pathways that are activated upon the action of cyclical tensile strains in pathophysiological conditions. In order to identify mechanosensors of tensile stresses on valvular interstitial cells, we employed biaxial cyclic strain of valvular cells in culture and quantitatively evaluated the expression of cell membrane proteins using a targeted protein array and interactome analyses. This approach yielded a high-throughput screening of all cell surface proteins involved in sensing mechanical stimuli. In this study, we were able to identify the cell membrane proteins which are activated during physiological cyclic tensile stresses of valvular cells. The proteins identified in this study were clustered into four interactomes, which included CC chemokine ligands, thrombospondin (adhesive glycoproteins), growth factors, and interleukins. The expression levels of these proteins generally indicated that cells tend to increase adhesive efforts to counteract the action of mechanical forces. This is the first study of this kind used to comprehensively identify the mechanosensitive proteins in valvular cells., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
35. On the Functional Role of Valve Interstitial Cell Stress Fibers: A Continuum Modeling Approach.
- Author
-
Sakamoto Y, Buchanan RM, Sanchez-Adams J, Guilak F, and Sacks MS
- Subjects
- Animals, Computer Simulation, Humans, Molecular Motor Proteins physiology, Stress, Mechanical, Heart Valves cytology, Heart Valves physiology, Mechanotransduction, Cellular physiology, Models, Cardiovascular, Myocardial Contraction physiology, Stress Fibers physiology
- Abstract
The function of the heart valve interstitial cells (VICs) is intimately connected to heart valve tissue remodeling and repair, as well as the onset and progression of valvular pathological processes. There is yet only very limited knowledge and extant models for the complex three-dimensional VIC internal stress-bearing structures, the associated cell-level biomechanical behaviors, and how they change under varying activation levels. Importantly, VICs are known to exist and function within the highly dynamic valve tissue environment, including very high physiological loading rates. Yet we have no knowledge on how these factors affect VIC function. To this end, we extended our previous VIC computational continuum mechanics model (Sakamoto, et al., 2016, "On Intrinsic Stress Fiber Contractile Forces in Semilunar Heart Valve Interstitial Cells Using a Continuum Mixture Model," J. Mech. Behav. Biomed. Mater., 54(244-258)). to incorporate realistic stress-fiber geometries, force-length relations (Hill model for active contraction), explicit α-smooth muscle actin (α-SMA) and F-actin expression levels, and strain rate. Novel micro-indentation measurements were then performed using cytochalasin D (CytoD), variable KCl molar concentrations, both alone and with transforming growth factor β1 (TGF-β1) (which emulates certain valvular pathological processes) to explore how α-SMA and F-actin expression levels influenced stress fiber responses under quasi-static and physiological loading rates. Simulation results indicated that both F-actin and α-SMA contributed substantially to stress fiber force generation, with the highest activation state (90 mM KCL + TGF-β1) inducing the largest α-SMA levels and associated force generation. Validation was performed by comparisons to traction force microscopy studies, which showed very good agreement. Interestingly, only in the highest activation state was strain rate sensitivity observed, which was captured successfully in the simulations. These unique findings demonstrated that only VICs with high levels of αSMA expression exhibited significant viscoelastic effects. Implications of this study include greater insight into the functional role of α-SMA and F-actin in VIC stress fiber function, and the potential for strain rate-dependent effects in pathological states where high levels of α-SMA occur, which appear to be unique to the valvular cellular in vivo microenvironment.
- Published
- 2017
- Full Text
- View/download PDF
36. Complete Static Repopulation of Decellularized Porcine Tissues for Heart Valve Engineering: An in vitro Study.
- Author
-
Roosens A, Asadian M, De Geyter N, Somers P, and Cornelissen R
- Subjects
- Adipose Tissue cytology, Animals, Cells, Cultured, Endothelial Cells cytology, Heart Valves chemistry, Mesenchymal Stem Cells cytology, Pericardium chemistry, Stem Cells cytology, Swine, Bioprosthesis, Heart Valve Prosthesis, Heart Valves cytology, Pericardium cytology, Tissue Engineering methods, Tissue Scaffolds chemistry
- Abstract
To date, a completely in vitro repopulated tissue-engineered heart valve has not been developed. This study focused on sequentially seeding 2 cell populations onto porcine decellularized heart valve leaflets (HVL) and pericardia (PER) to obtain fully repopulated tissues. For repopulation of the interstitium, porcine valvular interstitial cells (VIC) and bone marrow-derived mesenchymal stem cells (BM-MSC) or adipose tissue-derived stem cells (ADSC) were used. In parallel, the culture medium was supplemented with ascorbic acid 2-phosphate (AA) and its effect on recolonization was investigated. Subsequently and in order to obtain an endothelial surface layer similar to those in native HVL, valvular endothelial cells (VEC) were seeded onto the scaffolds. It was shown that VIC efficiently recolonized HVL and partially also PER. On the other hand, stem cells only demonstrated limited or no subsurface cell infiltration of HVL and PER. Interestingly, the addition of AA increased the migratory capacity of both stem cell populations. However, this was more pronounced for BM-MSC, and recolonization of HVL appeared to be more efficient than that of PER tissue. VEC were demonstrated to generate a new endothelial layer on HVL and PER. However, scanning microscopy revealed that these endothelial cells were not allowed to fully spread onto PER. This study provided a proof of concept for the future generation of a bioactive tissue-engineered heart valve by showing that bioactive HVL could be generated in vitro within 14 days via complete repopulation of the interstitium with BM-MSC or VIC and subsequent generation of an entirely new endothelium., (© 2017 S. Karger AG, Basel.)
- Published
- 2017
- Full Text
- View/download PDF
37. Isolated effect of material stiffness on valvular interstitial cell differentiation.
- Author
-
Coombs KE, Leonard AT, Rush MN, Santistevan DA, and Hedberg-Dirk EL
- Subjects
- Animals, Cells, Cultured, Heart Valves cytology, Osteoblasts cytology, Swine, Antigens, Differentiation biosynthesis, Cell Differentiation, Heart Valves metabolism, Methacrylates chemistry, Osteoblasts metabolism
- Abstract
Previous methods for investigating material stiffness on cell behavior have focused on the use of substrates with limited ranges of stiffness and/or fluctuating surface chemistries. Using the co-polymer system of n-octyl methacrylate crosslinked with diethylene glycol dimethacrylate (DEGDMA/nOM), we developed a new cell culture platform to analyze the isolated effects of stiffness independent from changes in surface chemistry. Materials ranging from 25 kPa to 4,700 kPa were fabricated. Surface analysis including goiniometry and X-ray photoelectron spectroscopy (XPS) confirmed consistent surface chemistry across all formulations examined. The mechanosensitive cell type valvular interstitial cell (VIC) was cultured DEGDMA/nOM substrates of differing stiffness. Results indicate that order of magnitude changes in stiffness do not increase gene expression of VIC alpha-smooth muscle actin (αSMA). However, structural organization of αSMA is altered on stiffer substrates, corresponding with the appearance of the osteoblastic marker osteocalcin and nodule formation. This research presents the co-polymer DEGDMA/nOM as ideal substrate to investigate the influence of stiffness on VIC differentiation without the confounding effects of changing material surface chemistry. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 51-61, 2017., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
38. Valve interstitial cell contractile strength and metabolic state are dependent on its shape.
- Author
-
Lam NT, Muldoon TJ, Quinn KP, Rajaram N, and Balachandran K
- Subjects
- Cell Nucleus ultrastructure, Cell Size, Cells, Cultured, Cytoskeleton ultrastructure, Humans, Shear Strength, Stress, Mechanical, Cell Nucleus physiology, Cytoskeleton physiology, Energy Metabolism physiology, Heart Valves cytology, Heart Valves physiology, Muscle Contraction physiology
- Abstract
The role of valvular interstitial cell (VIC) architecture in regulating cardiac valve function and pathology is not well understood. VICs are known to be more elongated in a hypertensive environment compared to those in a normotensive environment. We have previously reported that valve tissues cultured under hypertensive conditions are prone to acute pathological alterations in cell phenotype and contractility. We therefore aimed to rigorously study the relationship between VIC shape, contractile output and other functional indicators of VIC pathology. We developed an in vitro model to engineer VICs to take on the same shapes as those seen in normal and hypertensive conditions. VICs with longer cellular and nuclear shapes, as seen in hypertensive conditions, had greater contractile response to endothelin-1 that correlated with increased anisotropy of the actin architecture. These elongated VICs also demonstrated altered cell metabolism through a decreased optical redox ratio, which coincided with increased cellular proliferation. In the presence of actin polymerization inhibitor, however, these functional responses were significantly reduced, suggesting the important role of cytoskeletal actin organization in regulating cellular responses to abnormal shape. Overall, these results demonstrate the relationship between cell shape, cytoskeletal and nuclear organization, with functional output including contractility, metabolism, and proliferation.
- Published
- 2016
- Full Text
- View/download PDF
39. Valve interstitial cell shape modulates cell contractility independent of cell phenotype.
- Author
-
Tandon I, Razavi A, Ravishankar P, Walker A, Sturdivant NM, Lam NT, Wolchok JC, and Balachandran K
- Subjects
- Animals, Biomarkers metabolism, Cells, Cultured, Gene Expression Regulation, Heart Valves physiology, Muscle Contraction physiology, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle metabolism, Osteogenesis, Phenotype, Swine, Cell Shape, Heart Valves cytology
- Abstract
Valve interstitial cells are dispersed throughout the heart valve and play an important role in maintaining its integrity, function, and phenotype. While prior studies have detailed the role of external mechanical and biological factors in the function of the interstitial cell, the role of cell shape in regulating contractile function, in the context of normal and diseased phenotypes, is not well understood. Thus, the aim of this study was to elucidate the link between cell shape, phenotype, and acute functional contractile output. Valve interstitial cell monolayers with defined cellular shapes were engineered via constraining cells to micropatterned protein lines (10, 20, 40, 60 or 80µm wide). Samples were cultured in either normal or osteogenic medium. Cellular shape and architecture were quantified via fluorescent imaging techniques. Cellular contractility was quantified using a valve thin film assay and phenotype analyzed via western blotting, zymography, and qRT-PCR. In all pattern widths, cells were highly aligned, with maximum cell and nuclear elongation occurring for the 10μm pattern width. Cellular contractility was highest for the most elongated cells, but was also increased in cells on the widest pattern (80μm) that also had increased CX43 expression, suggesting a role for both elongated shape and increased cell-cell contact in regulating contractility. Cells cultured in osteogenic medium had greater expression of smooth muscle markers and correspondingly increased contractile stress responses. Cell phenotype did not significantly correlate with altered cell shape, suggesting that cellular shape plays a significant role in the regulation of valve contractile function independent of phenotype., (Copyright © 2016 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Role of cell-matrix interactions on VIC phenotype and tissue deposition in 3D PEG hydrogels.
- Author
-
Gould ST and Anseth KS
- Subjects
- Animals, Cells, Cultured, Heart Valves cytology, Swine, Collagen chemistry, Extracellular Matrix chemistry, Heart Valves metabolism, Hydrogels chemistry, Oligopeptides chemistry, Peptide Fragments chemistry, Polyethylene Glycols chemistry
- Abstract
Valvular interstitial cells (VICs) respond to 3D matrix interactions in a complex manner, but understanding these effects on VIC function better is important for applications ranging from valve tissue engineering to studying valve disease. Here, we encapsulated VICs in poly(ethylene glycol) (PEG) hydrogels modified with three different adhesive ligands, derived from fibronectin (RGDS), elastin (VGVAPG) and collagen-1 (P15). By day 14, VICs became significantly more elongated in RGDS-containing gels compared to VGVAPG or P15. This difference in cell morphology appeared to correlate with global matrix metalloproteinase (MMP) activity, as VICs encapsulated in RGDS-functionalized hydrogels secreted higher levels of active MMP at day 2. VIC activation to a myofibroblast phenotype was also characterized by staining for α-smooth muscle actin (αSMA) at day 14. The percentage of αSMA
+ VICs in the VGVAPG gels was the highest (56%) compared to RGDS (33%) or P15 (38%) gels. Matrix deposition and composition were also characterized at days 14 and 42 and found to depend on the initial hydrogel composition. All gel formulations had similar levels of collagen, elastin and chondroitin sulphate deposited as the porcine aortic valve. However, the composition of collagen deposited by VICs in VGVAPG-functionalized gels had a significantly higher collagen-X:collagen-1 ratio, which is associated with stenotic valves. Taken together, these data suggest that peptide-functionalized PEG hydrogels are a useful system for culturing VICs three-dimensionally and, with the ability to systematically alter biochemical and biophysical properties, this platform may prove useful in manipulating VIC function for valve regeneration. Copyright © 2013 John Wiley & Sons, Ltd., Competing Interests: Author Disclosure Statement: No competing financial interests exist., (Copyright © 2013 John Wiley & Sons, Ltd.)- Published
- 2016
- Full Text
- View/download PDF
41. Cells for tissue engineering of cardiac valves.
- Author
-
Jana S, Tranquillo RT, and Lerman A
- Subjects
- Animals, Heart Valves metabolism, Humans, Bioprosthesis, Heart Valve Prosthesis, Heart Valves cytology, Tissue Engineering methods
- Abstract
Heart valve tissue engineering is a promising alternative to prostheses for the replacement of diseased or damaged heart valves, because tissue-engineered valves have the ability to remodel, regenerate and grow. To engineer heart valves, cells are harvested, seeded onto or into a three-dimensional (3D) matrix platform to generate a tissue-engineered construct in vitro, and then implanted into a patient's body. Successful engineering of heart valves requires a thorough understanding of the different types of cells that can be used to obtain the essential phenotypes that are expressed in native heart valves. This article reviews different cell types that have been used in heart valve engineering, cell sources for harvesting, phenotypic expression in constructs and suitability in heart valve tissue engineering. Natural and synthetic biomaterials that have been applied as scaffold systems or cell-delivery platforms are discussed with each cell type. Copyright © 2015 John Wiley & Sons, Ltd., (Copyright © 2015 John Wiley & Sons, Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
42. Micro and nanotechnologies in heart valve tissue engineering.
- Author
-
Hasan A, Saliba J, Pezeshgi Modarres H, Bakhaty A, Nasajpour A, Mofrad MRK, and Sanati-Nezhad A
- Subjects
- Equipment Design, Heart Valves cytology, Humans, Male, Technology Assessment, Biomedical, Tissue Engineering methods, Bioartificial Organs, Heart Valves growth & development, Lab-On-A-Chip Devices, Nanoparticles chemistry, Organ Culture Techniques instrumentation, Tissue Engineering instrumentation, Tissue Scaffolds
- Abstract
Due to the increased morbidity and mortality resulting from heart valve diseases, there is a growing demand for off-the-shelf implantable tissue engineered heart valves (TEHVs). Despite the significant progress in recent years in improving the design and performance of TEHV constructs, viable and functional human implantable TEHV constructs have remained elusive. The recent advances in micro and nanoscale technologies including the microfabrication, nano-microfiber based scaffolds preparation, 3D cell encapsulated hydrogels preparation, microfluidic, micro-bioreactors, nano-microscale biosensors as well as the computational methods and models for simulation of biological tissues have increased the potential for realizing viable, functional and implantable TEHV constructs. In this review, we aim to present an overview of the importance and recent advances in micro and nano-scale technologies for the development of TEHV constructs., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. Pediatric cardiovascular grafts: historical perspective and future directions.
- Author
-
Heydrick S, Roberts E, Kim J, Emani S, and Wong JY
- Subjects
- Heart Valve Prosthesis, Humans, Cardiovascular Diseases therapy, Fetal Stem Cells cytology, Heart Valves cytology, Tissue Engineering methods
- Abstract
Tissue-engineered cardiovascular patches, cardiac valves, and great vessels are emerging solutions for the surgical treatment of congenital cardiovascular abnormalities due to their potential for adapting with the growing child. The ideal pediatric cardiovascular patch/graft is non-thrombogenic, phenotypically compatible, and matches the compliance and mechanical strength of the native tissue, both initially and throughout growth. Bottom-up tissue engineering approaches, in which three-dimensional tissue is built layer-by-layer from scaffold-less cell sheets in vitro, offer an exciting potential solution. Cell source variability, sheet patterning, and scaffold-less fabrication are promising advantages offered by this approach. Here we review the latest developments and next steps in bottom-up tissue engineering targeted at meeting the necessary design criteria for successful pediatric cardiac tissue-engineered grafts., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. High-throughput investigation of endothelial-to-mesenchymal transformation (EndMT) with combinatorial cellular microarrays.
- Author
-
Wang Z, Calpe B, Zerdani J, Lee Y, Oh J, Bae H, Khademhosseini A, and Kim K
- Subjects
- Animals, Extracellular Matrix Proteins analysis, Extracellular Matrix Proteins genetics, Extracellular Matrix Proteins metabolism, Heart Valves cytology, Intercellular Signaling Peptides and Proteins analysis, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Rats, Swine, Cell Transdifferentiation physiology, Endothelial Cells cytology, Endothelial Cells metabolism, High-Throughput Screening Assays methods, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Tissue Array Analysis methods
- Abstract
In the developing heart, a specific subset of endocardium undergoes an endothelial-to-mesenchymal transformation (EndMT) thus forming nascent valve leaflets. Extracellular matrix (ECM) proteins and growth factors (GFs) play important roles in regulating EndMT but the combinatorial effect of GFs with ECM proteins is less well understood. Here we use microscale engineering techniques to create single, binary, and tertiary component microenvironments to investigate the combinatorial effects of ECM proteins and GFs on the attachment and transformation of adult ovine mitral valve endothelial cells to a mesenchymal phenotype. With the combinatorial microenvironment microarrays, we utilized 60 different combinations of ECM proteins (Fibronectin, Collagen I, II, IV, Laminin) and GFs (TGF-β1, bFGF, VEGF) and were able to identify new microenvironmental conditions capable of modulating EndMT in MVECs. Experimental results indicated that TGF-β1 significantly upregulated the EndMT while either bFGF or VEGF downregulated EndMT process markedly. Also, ECM proteins could influence both the attachment of MVECs and the response of MVECs to GFs. In terms of attachment, fibronectin is significantly better for the adhesion of MVECs among the five tested proteins. Overall collagen IV and fibronectin appeared to play important roles in promoting EndMT process. Great consistency between macroscale and microarrayed experiments and present studies demonstrates that high-throughput cellular microarrays are a promising approach to study the regulation of EndMT in valvular endothelium. Biotechnol. Bioeng. 2016;113: 1403-1412. © 2015 Wiley Periodicals, Inc., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
45. Real-time 3D visualization of cellular rearrangements during cardiac valve formation.
- Author
-
Pestel J, Ramadass R, Gauvrit S, Helker C, Herzog W, and Stainier DY
- Subjects
- Animals, Cell Movement, Coronary Circulation, Endocardium cytology, Endocardium embryology, Gene Expression Regulation, Developmental, Heart Atria cytology, Heart Atria embryology, Heart Ventricles cytology, Heart Ventricles embryology, Mutation genetics, Myocardial Contraction, Organogenesis genetics, Receptors, Notch metabolism, Wnt Signaling Pathway, Zebrafish, Heart Valves cytology, Heart Valves embryology, Imaging, Three-Dimensional
- Abstract
During cardiac valve development, the single-layered endocardial sheet at the atrioventricular canal (AVC) is remodeled into multilayered immature valve leaflets. Most of our knowledge about this process comes from examining fixed samples that do not allow a real-time appreciation of the intricacies of valve formation. Here, we exploit non-invasive in vivo imaging techniques to identify the dynamic cell behaviors that lead to the formation of the immature valve leaflets. We find that in zebrafish, the valve leaflets consist of two sets of endocardial cells at the luminal and abluminal side, which we refer to as luminal cells (LCs) and abluminal cells (ALCs), respectively. By analyzing cellular rearrangements during valve formation, we observed that the LCs and ALCs originate from the atrium and ventricle, respectively. Furthermore, we utilized Wnt/β-catenin and Notch signaling reporter lines to distinguish between the LCs and ALCs, and also found that cardiac contractility and/or blood flow is necessary for the endocardial expression of these signaling reporters. Thus, our 3D analyses of cardiac valve formation in zebrafish provide fundamental insights into the cellular rearrangements underlying this process., (© 2016. Published by The Company of Biologists Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
46. Relative Effects of Fluid Oscillations and Nutrient Transport in the In Vitro Growth of Valvular Tissues.
- Author
-
Salinas M, Rath S, Villegas A, Unnikrishnan V, and Ramaswamy S
- Subjects
- Animals, Biomarkers analysis, Biomarkers metabolism, Cells, Cultured, Collagen analysis, Collagen metabolism, Gene Expression Profiling, Glucose metabolism, Hydrodynamics, Oxygen metabolism, Porosity, Sheep, Bioreactors, Heart Valves chemistry, Heart Valves cytology, Heart Valves metabolism, Heart Valves physiology, Tissue Engineering methods
- Abstract
Engineered valvular tissues are cultured dynamically, and involve specimen movement. We previously demonstrated that oscillatory shear stresses (OSS) under combined steady flow and specimen cyclic flexure (flex-flow) promote tissue formation. However, localized efficiency of specimen mass transport is also important in the context of cell viability within the growing tissues. Here, we investigated the delivery of two essential species for cell survival, glucose and oxygen, to 3-dimensional (3D) engineered valvular tissues. We applied a convective-diffusive model to characterize glucose and oxygen mass transport with and without valve-like specimen flexural movement. We found the mass transport effects for glucose and oxygen to be negligible for scaffold porosities typically present during in vitro experiments and non-essential unless the porosity was unusually low (<40%). For more typical scaffold porosities (75%) however, we found negligible variation in the specimen mass fraction of glucose and oxygen in both non-moving and moving constructs (p > 0.05). Based on this result, we conducted an experiment using bone marrow stem cell (BMSC)-seeded scaffolds under Pulsatile flow-alone states to permit OSS without any specimen movement. BMSC-seeded specimen collagen from the pulsatile flow and flex-flow environments were subsequently found to be comparable (p > 0.05) and exhibited some gene expression similarities. We conclude that a critical magnitude of fluid-induced, OSS created by either pulsatile flow or flex-flow conditions, particularly when the oscillations are physiologically-relevant, is the direct, principal stimulus that promotes engineered valvular tissues and its phenotype, whereas mass transport benefits derived from specimen movement are minimal.
- Published
- 2016
- Full Text
- View/download PDF
47. Hyaluronan Hydrogels for a Biomimetic Spongiosa Layer of Tissue Engineered Heart Valve Scaffolds.
- Author
-
Puperi DS, O'Connell RW, Punske ZE, Wu Y, West JL, and Grande-Allen KJ
- Subjects
- Animals, Cells, Cultured, Proteoglycans, Swine, Tensile Strength, Biomimetics methods, Heart Valves cytology, Hyaluronic Acid chemistry, Hydrogels chemistry, Tissue Engineering methods, Tissue Scaffolds
- Abstract
Advanced tissue engineered heart valves must be constructed from multiple materials to better mimic the heterogeneity found in the native valve. The trilayered structure of aortic valves provides the ability to open and close consistently over a full human lifetime, with each layer performing specific mechanical functions. The middle spongiosa layer consists primarily of proteoglycans and glycosaminoglycans, providing lubrication and dampening functions as the valve leaflet flexes open and closed. In this study, hyaluronan hydrogels were tuned to perform the mechanical functions of the spongiosa layer, provide a biomimetic scaffold in which valve cells were encapsulated in 3D for tissue engineering applications, and gain insight into how valve cells maintain hyaluronan homeostasis within heart valves. Expression of the HAS1 isoform of hyaluronan synthase was significantly higher in hyaluronan hydrogels compared to blank-slate poly(ethylene glycol) diacrylate (PEGDA) hydrogels. Hyaluronidase and matrix metalloproteinase enzyme activity was similar between hyaluronan and PEGDA hydrogels, even though these scaffold materials were each specifically susceptible to degradation by different enzyme types. KIAA1199 was expressed by valve cells and may play a role in the regulation of hyaluronan in heart valves. Cross-linked hyaluronan hydrogels maintained healthy phenotype of valve cells in 3D culture and were tuned to approximate the mechanical properties of the valve spongiosa layer. Therefore, hyaluronan can be used as an appropriate material for the spongiosa layer of a proposed laminate tissue engineered heart valve scaffold.
- Published
- 2016
- Full Text
- View/download PDF
48. Customized Interface Biofunctionalization of Decellularized Extracellular Matrix: Toward Enhanced Endothelialization.
- Author
-
Aubin H, Mas-Moruno C, Iijima M, Schütterle N, Steinbrink M, Assmann A, Gil FJ, Lichtenberg A, Pegueroles M, and Akhyari P
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cells, Cultured, Humans, Male, Rats, Rats, Wistar, Sheep, Tissue Scaffolds chemistry, Cell Adhesion physiology, Extracellular Matrix chemistry, Heart Valves cytology, Human Umbilical Vein Endothelial Cells cytology, Pulmonary Valve cytology, Tissue Engineering methods
- Abstract
Interface biofunctionalization strategies try to enhance and control the interaction between implants and host organism. Decellularized extracellular matrix (dECM) is widely used as a platform for bioengineering of medical implants, having shown its suitability in a variety of preclinical as well as clinical models. In this study, specifically designed, custom-made synthetic peptides were used to functionalize dECM with different cell adhesive sequences (RGD, REDV, and YIGSR). Effects on in vitro endothelial cell adhesion and in vivo endothelialization were evaluated in standardized models using decellularized ovine pulmonary heart valve cusps (dPVCs) and decellularized aortic grafts (dAoGs), respectively. Contact angle measurements and fluorescent labeling of custom-made peptides showed successful functionalization of dPVCs and dAoGs. The functionalization of dPVCs with a combination of bioactive sequences significantly increased in vitro human umbilical vein endothelial cell adhesion compared to nonfunctionalized controls. In a functional rodent aortic transplantation model, fluorescent-labeled peptides on dAoGs were persistent up to 10 days in vivo under exposure to systemic circulation. Although there was a trend toward enhanced in vivo endothelialization of functionalized grafts compared to nonfunctionalized controls, there was no statistical significance and a large biological variability in both groups. Despite failing to show a clear biological effect in the used in vivo model system, our initial findings do suggest that endothelialization onto dECM may be modulated by customized interface biofunctionalization using the presented method. Since bioactive sequences within the dECM-synthetic peptide platform are easily interchangeable and combinable, further control of host cell proliferation, function, and differentiation seems to be feasible, possibly paving the way to a new generation of multifunctional dECM scaffolds for regenerative medicine.
- Published
- 2016
- Full Text
- View/download PDF
49. [Effect of rapamycin on proliferation of rat heart valve interstitial cells in vitro].
- Author
-
Tan Y, Wang JY, Yi RL, and Qiu J
- Subjects
- Animals, Blotting, Western, Cell Cycle, Cells, Cultured, Phosphorylation, Rats, Cell Proliferation drug effects, Heart Valves cytology, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Sirolimus pharmacology
- Abstract
Objective: To investigate the effect of rapamycin on the proliferation of rat valvular interstitial cells in primary culture., Methods: The interstitial cells isolated from rat aortic valves were cultured and treated with rapamycin, and the cell growth and cell cycle changes were analyzed using MTT assay and flow cytometry, respectively. RT-PCR was used to detect mRNA expression levels of S6 and P70S6K in cells, and the protein expressions level of S6, P70S6K, P-S6, and P-P70S6K were detected using Western blotting., Results: Rat aortic valvular interstitial cells was isolated successfully. The rapamycin-treated cells showed a suppressed proliferative activity (P<0.05), but the cell cycle distribution remained unaffected. Rapamycin treatment resulted in significantly decreased S6 and P70S6K protein phosphorylation level in the cells (P<0.05)., Conclusion: The mechanism by which rapamycin inhibits the proliferation of valvular interstitial cells probably involves suppression of mTOR to lower S6 and P70S6K phosphorylation level but not direct regulation of the cell cycle.
- Published
- 2016
50. Endocardial-to-mesenchymal transformation and mesenchymal cell colonization at the onset of human cardiac valve development.
- Author
-
Monaghan MG, Linneweh M, Liebscher S, Van Handel B, Layland SL, and Schenke-Layland K
- Subjects
- Cell Adhesion Molecules metabolism, Cell Count, Cell Differentiation, Cell Proliferation, Endothelial Cells metabolism, Female, Gene Expression Regulation, Developmental, Humans, Hyaluronan Receptors metabolism, NFATC Transcription Factors genetics, NFATC Transcription Factors metabolism, Pregnancy, Pregnancy Trimester, Second, Spatio-Temporal Analysis, Time Factors, Vascular Endothelial Growth Factor A metabolism, Endocardium embryology, Heart Valves cytology, Heart Valves embryology, Mesoderm cytology, Mesoderm embryology
- Abstract
The elucidation of mechanisms in semilunar valve development might enable the development of new therapies for congenital heart disorders. Here, we found differences in proliferation-associated genes and genes repressed by VEGF between human semilunar valve leaflets from first and second trimester hearts. The proliferation of valve interstitial cells and ventricular valve endothelial cells (VECs) and cellular density declined from the first to the second trimester. Cytoplasmic expression of NFATC1 was detected in VECs (4 weeks) and, later, cells in the leaflet/annulus junction mesenchyme expressing inactive NFATC1 (5.5-9 weeks) were detected, indicative of endocardial-to-mesenchymal transformation (EndMT) in valvulogenesis. At this leaflet/annulus junction, CD44(+) cells clustered during elongation (11 weeks), extending toward the tip along the fibrosal layer in second trimester leaflets. Differing patterns of maturation in the fibrosa and ventricularis were detected via increased fibrosal periostin content, which tracked the presence of the CD44(+) cells in the second trimester. We revealed that spatiotemporal NFATC1 expression actively regulates EndMT during human valvulogenesis, as early as 4 weeks. Additionally, CD44(+) cells play a role in leaflet maturation toward the trilaminar structure, possibly via migration of VECs undergoing EndMT, which subsequently ascend from the leaflet/annulus junction., (© 2016. Published by The Company of Biologists Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.