12 research outputs found

    Spheroid three-dimensional culture enhances Notch signaling in cardiac progenitor cells

    Get PDF
    Cardiac progenitor cells (CPCs) are a promising candidate for cardiac regeneration, and the interaction between CPCs and their microenvironment can influence their regenerative response. Notch signaling plays a key role in cell fate decisions in the developing and adult heart. Here, we investigated the effect of three-dimensional (3D) spheroid culture, as a model of the 3D microenvironment, on Notch in fetal and adult human CPCs, under room air (20%) and physiological (5%) oxygen tension. Notch signaling is enhanced in 3D spheroids; spheroid culture under 5% O2 further increases Notch signaling enhancement, and might ultimately improve the regenerative potential of CPCs

    The interplay between cardiac progenitor cells and their microenvironment

    No full text

    Tissue engineering for skeletal muscle regeneration

    Get PDF
    Stem cells and regenerative medicine have obtained a remarkable consent from the scientific community for their promising ability to recover aged, injured and diseased tissue. However, despite the noteworthy potential, hurdles currently hinder their use and clinical application: cell survival, immune response, tissue engraftment and efficient differentiation. Hence a new interdisciplinary scientific approach, such as tissue engineering, is going deep attempts to mimic neo-tissue-genesis as well as stem cell engraftment amelioration. Skeletal muscle tissue engineering represents a great potentiality in medicine for muscle regeneration exploiting new generation injectable hydrogel as scaffold supporting progenitor/stem cells for muscle differentiation reconstructing the natural skeletal muscle tissue architecture influenced by matrix mechanical and physical property and by a dynamic environmen

    Cardiac progenitor cells and the interplay with their microenvironment

    Get PDF
    The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells,\u3cbr/\u3eextracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity

    Cardiomyocyte progenitor cell mechanoresponse unrevealed: strain avoidance and mechanosome development.

    No full text
    For emerging cardiac regeneration strategies, it is essential to know if and how cardiac stem cells sense and respond to the mechanical stimuli provided by their environment in the beating heart. Here, we study the response to cyclic strain of undifferentiated and predifferentiated human cardiomyocyte progenitor cells (CMPCs), as well as the formation and activation of the cellular structures involved in mechanosensing, that we termed 'mechanosome'. Once verified that the applied uniaxial cyclic strain (10%, 0.5 Hz) did not alter the cardiac lineage commitment and differentiation state of CMPCs, the cellular mechanoresponse to the applied strain was quantified by cellular orientation. While undifferentiated cells maintained their original (random) orientation, upon early cardiomyogenic differentiation (predifferentiated) CMPCs exhibited a distinct strain avoidance response after 48 h of cyclic straining. Interestingly, the mechanosome development and the activation of the mechanotransduction pathways also occurred with early cardiac differentiation of the CMPCs, regardless of the substrate or the applied cyclic strain. These results indicate that the mechanoresponse of CMPCs depends on the presence of a developed mechanosome, which only develops during early cardiomyogenic differentiation Our findings provide the first understanding of mechanotransduction in human CMPCs and as such can contribute to the improvement of cardiac regeneration strategies

    Behavior of CMPCs in unidirectional constrained and stress-free 3D hydrogels

    No full text
    Cardiomyocyte progenitor cells (CMPCs) are a candidate cell source for cardiac regenerative therapy. However, like other stem cells, after transplantation in the heart, cell retention and differentiation capacity of the CMPCs are low. Combining cells with biomaterials might overcome this problem. By serving as a (temporal) environment, the biomaterial can retain the cells and provide signals that enhance survival, proliferation and differentiation of the cells. To gain more insight into the effect that the encapsulation of CMPCs in a biomaterial has on their behavior, we cultured CMPCs in unidirectional constrained and stress-free collagen/Matrigel hydrogels. CMPCs cultured in 3D hydrogels stay viable and keep their cardiomyogenic profile independent of the application of strain. Moreover, the increased expression of Nkx2.5, myocardin and cTnT in 3D hydrogels compared to 2D cultures, suggests enhanced cardiomyogenic differentiation capacity of cells in 3D. Furthermore, increased expression of collagen I, collagen III, elastin and fibronectin and of the matrix remodeling enzymes MMP-1, MMP-2, MMP-9, and TIMP-1 and TIMP-2 in the 3D hydrogels is indicative of an enhanced matrix remodeling capacity of CMPCs in a 3D environment, independent of the application of strain. Interestingly, the additional application of static strain to the 3D hydrogels, as imposed by hydrogel constrainment, stabilized CMPC viability and proliferation, resulted in enhanced cardiac marker protein expression and appeared crucial for cellular organization and morphology. More specifically, CMPCs cultured in 3D collagen/Matrigel constrained hydrogels became readily mechanosensitive, had a rod-shaped morphology, and responded to the applied strain by orienting in the direction of the constraint. Overall, our data demonstrate the applicability of CMPCs in a 3D environment since encapsulation of CMPCs may stabilize survival and proliferation, can enhance the differentiation and remodeling capacity of the cells, and could induce cellular re-organization, which all may contribute to an improved efficiency of cardiac stem cell therapy

    Spheroid three-dimensional culture enhances Notch signaling in cardiac progenitor cells

    No full text
    \u3cp\u3eCardiac progenitor cells (CPCs) are a promising candidate for cardiac regeneration, and the interaction between CPCs and their microenvironment can influence their regenerative response. Notch signaling plays a key role in cell fate decisions in the developing and adult heart. Here, we investigated the effect of three-dimensional (3D) spheroid culture, as a model of the 3D microenvironment, on Notch in fetal and adult human CPCs, under room air (20%) and physiological (5%) oxygen tension. Notch signaling is enhanced in 3D spheroids; spheroid culture under 5% O\u3csub\u3e2\u3c/sub\u3e further increases Notch signaling enhancement, and might ultimately improve the regenerative potential of CPCs.\u3c/p\u3
    corecore