scholarly journals Advancing cardiovascular tissue engineering

F1000Research ◽  
2016 ◽  
Vol 5 ◽  
pp. 1045 ◽  
Author(s):  
George A. Truskey

Cardiovascular tissue engineering offers the promise of biologically based repair of injured and damaged blood vessels, valves, and cardiac tissue. Major advances in cardiovascular tissue engineering over the past few years involve improved methods to promote the establishment and differentiation of induced pluripotent stem cells (iPSCs), scaffolds from decellularized tissue that may produce more highly differentiated tissues and advance clinical translation, improved methods to promote vascularization, and novel in vitro microphysiological systems to model normal and diseased tissue function. iPSC technology holds great promise, but robust methods are needed to further promote differentiation. Differentiation can be further enhanced with chemical, electrical, or mechanical stimuli.

ASAIO Journal ◽  
2002 ◽  
Vol 48 (1) ◽  
pp. 8-11 ◽  
Author(s):  
Stefan Jockenhoevel ◽  
Gregor Zund ◽  
Simon P. Hoerstrup ◽  
Andrea Schnell ◽  
Marco Turina

2021 ◽  
Vol 16 (12) ◽  
pp. 1037-1050
Author(s):  
Tahmineh Kazemi ◽  
Ahmad A Mohammadpour ◽  
Maryam M Matin ◽  
Nasser Mahdavi-Shahri ◽  
Hesam Dehghani ◽  
...  

Aim: To evaluate the suitability of using aorta elastin scaffold, in combination with human adipose-derived mesenchymal stem cells (hAd-MSCs), as an approach for cardiovascular tissue engineering. Materials & Methods: Human adipose-derived MSCs were seeded on elastin samples of decellularized bovine aorta. The samples were cultured in vitro to investigate the inductive effects of this scaffold on the cells. The results were evaluated using histological, and immunohistochemical methods, as well as MTT assay, DNA content, reverse transcription-PCR and scanning electron microscopy. Results: Histological staining and DNA content confirmed the efficacy of decellularization procedure (82% DNA removal). MTT assay showed the construct’s ability to support cell viability and proliferation. Cell differentiation was confirmed by reverse transcription-PCR and positive immunohistochemistry for alfa smooth muscle actin and von Willebrand. Conclusion: The prepared aortic elastin samples act as a potential scaffold, in combination with MSCs, for applications in cardiovascular tissue engineering. Further experiments in animal models are required to confirm this.


2010 ◽  
Vol 109 (5) ◽  
pp. 1483-1491 ◽  
Author(s):  
Marijke A. A. van Vlimmeren ◽  
Anita Driessen-Mol ◽  
Marloes van den Broek ◽  
Carlijn V. C. Bouten ◽  
Frank P. T. Baaijens

In vivo functionality of cardiovascular tissue engineered constructs requires in vitro control of tissue development to obtain a well developed extracellular matrix (ECM). We hypothesize that ECM formation and maturation is stimulated by culturing at low oxygen concentrations. Gene expression levels of monolayers of human vascular-derived myofibroblasts, exposed to 7, 4, 2, 1, and 0.5% O2( n = 9 per group) for 24 h, were measured for vascular endothelial growth factor (VEGF), procollagen α1(I) and α1(III), elastin, and cross-link enzymes lysyl oxidase (LOX) and lysyl hydroxylase 2 (LH2). After 4 days of exposure to 7, 2, and 0.5% O2( n = 3 per group), protein synthesis was evaluated. All analyses were compared with control cultures at 21% O2. Human myofibroblasts turned to hypoxia-driven gene expression, indicated by VEGF expression, at oxygen concentrations of 4% and lower. Gene expression levels of procollagen α1(I) and α1(III) increased to 138 ± 26 and 143 ± 19%, respectively, for all oxygen concentrations below 4%. At 2% O2, LH2 and LOX gene expression levels were higher than control cultures (340 ± 53 and 136 ± 29%, respectively), and these levels increased even further with decreasing oxygen concentrations (611 ± 176 and 228 ± 45%, respectively, at 0.5% O2). Elastin gene expression levels remained unaffected. Collagen synthesis and LH2 protein levels increased at oxygen concentrations of 2% and lower. Oxygen concentrations below 4% induce enhanced ECM production by human myofibroblasts. Implementation of these results in cardiovascular tissue engineering approaches enables in vitro control of tissue development.


Author(s):  
Jia Min Lee ◽  
Swee Leong Sing ◽  
Edgar Yong Sheng Tan ◽  
Wai Yee Yeong

Fabrication techniques for cardiac tissue engineering have been evolving around scaffold-based and scaf-fold-free approaches. Conventional fabrication approaches lack control over scalability and homogeneous cell distribu-tion. Bioprinting provides a technological platform for controlled deposition of biomaterials, cells, and biological fac-tors in an organized fashion. Bioprinting is capable of alternating heterogeneous cell printing, printing anatomical rele-vant structure and microchannels resembling vasculature network. These are essential features of an engineered cardiac tissue. Bioprinting can potentially build engineered cardiac construct that resembles native tissue across macro to na-noscale.


2021 ◽  
Vol 8 (11) ◽  
pp. 137
Author(s):  
Astha Khanna ◽  
Maedeh Zamani ◽  
Ngan F. Huang

Regenerative medicine and tissue engineering strategies have made remarkable progress in remodeling, replacing, and regenerating damaged cardiovascular tissues. The design of three-dimensional (3D) scaffolds with appropriate biochemical and mechanical characteristics is critical for engineering tissue-engineered replacements. The extracellular matrix (ECM) is a dynamic scaffolding structure characterized by tissue-specific biochemical, biophysical, and mechanical properties that modulates cellular behavior and activates highly regulated signaling pathways. In light of technological advancements, biomaterial-based scaffolds have been developed that better mimic physiological ECM properties, provide signaling cues that modulate cellular behavior, and form functional tissues and organs. In this review, we summarize the in vitro, pre-clinical, and clinical research models that have been employed in the design of ECM-based biomaterials for cardiovascular regenerative medicine. We highlight the research advancements in the incorporation of ECM components into biomaterial-based scaffolds, the engineering of increasingly complex structures using biofabrication and spatial patterning techniques, the regulation of ECMs on vascular differentiation and function, and the translation of ECM-based scaffolds for vascular graft applications. Finally, we discuss the challenges, future perspectives, and directions in the design of next-generation ECM-based biomaterials for cardiovascular tissue engineering and clinical translation.


Author(s):  
Alex Savchenko ◽  
Rose T. Yin ◽  
Dmitry Kireev ◽  
Igor R. Efimov ◽  
Elena Molokanova

Cardiac tissue engineering requires materials that can faithfully recapitulate and support the native in vivo microenvironment while providing a seamless bioelectronic interface. Current limitations of cell scaffolds include the lack of electrical conductivity and suboptimal mechanical properties. Here we discuss how the incorporation of graphene into cellular scaffolds, either alone or in combination with other materials, can affect morphology, function, and maturation of cardiac cells. We conclude that graphene-based scaffolds hold great promise for cardiac tissue engineering.


2016 ◽  
Vol 4 (9) ◽  
pp. 1361-1370 ◽  
Author(s):  
Israel Gonzalez de Torre ◽  
Miriam Weber ◽  
Luis Quintanilla ◽  
Matilde Alonso ◽  
Stefan Jockenhoevel ◽  
...  

In the field of tissue engineering, the properties of the scaffolds are of crucial importance for the success of the application.


Author(s):  
Cansu Karakaya ◽  
Jordy G. M. van Asten ◽  
Tommaso Ristori ◽  
Cecilia M. Sahlgren ◽  
Sandra Loerakker

AbstractCardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell–cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell–cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell–cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell–cell signaling to highlight their potential role in future CVTE strategies.


Sign in / Sign up

Export Citation Format

Share Document