17 research outputs found

    Stress-vs-time signals allow the prediction of structurally catastrophic events during fracturing of immature cartilage and predetermine the biomechanical, biochemical, and structural impairment

    Get PDF
    Objective Trauma-associated cartilage fractures occur in children and adolescents with clinically significant incidence. Several studies investigated biomechanical injury by compressive forces but the injury-related stress has not been investigated extensively. In this study, we hypothesized that the biomechanical stress occurring during compressive injury predetermines the biomechanical, biochemical, and structural consequences. We specifically investigated whether the stress-vs-time signal correlated with the injurious damage and may allow prediction of cartilage matrix fracturing. Methods Superficial and deeper zones disks (SZDs, DZDs; immature bovine cartilage) were biomechanically characterized, injured (50% compression, 100%/s strain-rate), and re-characterized. Correlations of the quantified functional, biochemical and histological damage with biomechanical parameters were zonally investigated. Results Injured SZDs exhibited decreased dynamic stiffness (by 93.04 ± 1.72%), unresolvable equilibrium moduli, structural damage (2.0 ± 0.5 on a 5-point-damage-scale), and 1.78-fold increased sGAG loss. DZDs remained intact. Measured stress-vs-time-curves during injury displayed 4 distinct shapes, which correlated with histological damage (p < 0.001), loss of dynamic stiffness and sGAG (p < 0.05). Damage prediction in a blinded experiment using stress-vs-time grades was 100%-correct and sensitive to differentiate single/complex matrix disruptions. Correlations of the dissipated energy and maximum stress rise with the extent of biomechanical and biochemical damage reached significance when SZDs and DZDs were analyzed as zonal composites but not separately. Conclusions The biomechanical stress that occurs during compressive injury predetermines the biomechanical, biochemical, and structural consequences and, thus, the structural and functional damage during cartilage fracturing. A novel biomechanical method based on the interpretation of compressive yielding allows the accurate prediction of the extent of structural damage.National Institutes of Health (U.S.) (Grant R01-AR45779)Deutsche Forschungsgemeinschaft (Grant RO2511/1-1)Deutsche Forschungsgemeinschaft (Grant RO2511/2-1)Germany. Federal Ministry of Education and Research (Grant 01KQ0902B TP2

    The geometrical shape of mesenchymal stromal cells measured by quantitative shape descriptors is determined by the stiffness of the biomaterial and by cyclic tensile forces

    Full text link
    Controlling mesenchymal stromal cell (MSC) shape is a novel method for investigating and directing MSC behaviour in vitro. it was hypothesized that specifigc MSC shapes can be generated by using stiffnessâ defined biomaterial surfaces and by applying cyclic tensile forces. Biomaterials used were thin and thick silicone sheets, fibronectin coating, and compacted collagen type I sheets. The MSC morphology was quantified by shape descriptors describing dimensions and membrane protrusions. Nanoscale stiffness was measured by atomic force microscopy and the expression of smooth muscle cell (SMC) marker genes (ACTA2, TAGLN, CNN1) by quantitative reverseâ transcription polymerase chain reaction. Cyclic stretch was applied with 2.5% or 5% amplitudes. Attachment to biomaterials with a higher stiffness yielded more elongated MSCs with fewer membrane protrusions compared with biomaterials with a lower stiffness. For cyclic stretch, compacted collagen sheets were selected, which were associated with the most elongated MSC shape across all investigated biomaterials. As expected, cyclic stretch elongated MSCs during stretch. One hour after cessation of stretch, however, MSC shape was rounder again, suggesting loss of stretchâ induced shape. Different shape descriptor values obtained by different stretch regimes correlated significantly with the expression levels of SMC marker genes. Values of approximately 0.4 for roundness and 3.4 for aspect ratio were critical for the highest expression levels of ACTA2 and CNN1. Thus, specific shape descriptor values, which can be generated using biomaterialâ associated stiffness and tensile forces, can serve as a template for the induction of specific gene expression levels in MSC. Copyright © 2017 John Wiley & Sons, Ltd.Peer Reviewedhttps://deepblue.lib.umich.edu/bitstream/2027.42/141253/1/term2263.pdfhttps://deepblue.lib.umich.edu/bitstream/2027.42/141253/2/term2263_am.pd

    Smooth Muscle-Like Cells Generated from Human Mesenchymal Stromal Cells Display Marker Gene Expression and Electrophysiological Competence Comparable to Bladder Smooth Muscle Cells

    No full text
    The use of mesenchymal stromal cells (MSCs) differentiated toward a smooth muscle cell (SMC) phenotype may provide an alternative for investigators interested in regenerating urinary tract organs such as the bladder where autologous smooth muscle cells cannot be used or are unavailable. In this study we measured the effects of good manufacturing practice (GMP)-compliant expansion followed by myogenic differentiation of human MSCs on the expression of a range of contractile (from early to late) myogenic markers in relation to the electrophysiological parameters to assess the functional role of the differentiated MSCs and found that differentiation of MSCs associated with electrophysiological competence comparable to bladder SMCs. Within 1-2 weeks of myogenic differentiation, differentiating MSCs significantly expressed alpha smooth muscle actin (αSMA; ACTA2), transgelin (TAGLN), calponin (CNN1), and smooth muscle myosin heavy chain (SM-MHC; MYH11) according to qRT-PCR and/or immunofluorescence and Western blot. Voltage-gated Na+ current levels also increased within the same time period following myogenic differentiation. In contrast to undifferentiated MSCs, differentiated MSCs and bladder SMCs exhibited elevated cytosolic Ca2+ transients in response to K+-induced depolarization and contracted in response to K+ indicating functional maturation of differentiated MSCs. Depolarization was suppressed by Cd2+, an inhibitor of voltage-gated Ca2+-channels. The expression of Na+-channels was pharmacologically identified as the Nav1.4 subtype, while the K+ and Ca2+ ion channels were identified by gene expression of KCNMA1, CACNA1C and CACNA1H which encode for the large conductance Ca2+-activated K+ channel BKCa channels, Cav1.2 L-type Ca2+ channels and Cav3.2 T-type Ca2+ channels, respectively. This protocol may be used to differentiate adult MSCs into smooth muscle-like cells with an intermediate-to-late SMC contractile phenotype exhibiting voltage-gated ion channel activity comparable to bladder SMCs which may be important for urological regenerative medicine applications

    Expression levels of αSMA <i>ex vivo</i>, after MSCs expansion and myogenic differentiation <i>in vitro</i>.

    No full text
    <p>(A) Mononuclear cells were directly isolated from bone marrow <i>ex vivo</i> and assessed for expression of αSMA by flow cytometry. Viable cells were gated (SSC/FSC), CD45<sup>+</sup> cells were excluded, and expression of αSMA was recorded in the cytoplasm of the CD45<sup>-</sup>CD271<sup>+</sup> fraction of the mononuclear cells. The histogram represents αSMA in the cytoplasm of CD271<sup>+</sup> cells (solid line) compared to the unstained controls (dotted line). (B) MSCs were expanded in GMP-compliant expansion medium and then assessed for expression of αSMA in the cytoplasm of the cells. Viable cells were gated (SSC/FSC) and αSMA<sup>+</sup> cells were recorded (solid line). The dotted histogram represents the unstained controls. The broad profile of the histogram indicates that a large portion of MSCs express αSMA after expansion and prior to induction of differentiation (27% of MSCs were positive, MFI of 38). (C) After expansion in GMP-compliant expansion medium MSCs were differentiated for 14 days. Then expression of αSMA in the cytoplasm of differentiating MSCs was explored (solid line). The histogram indicates that more cells contain αSMA after differentiation (27% of MSCs were positive, MFI of 42, right panel). The dotted lines represent unstained controls.</p

    Expression of contractile SMC-specific proteins analyzed by immunofluorescence.

    No full text
    <p>MSCs were expanded in GMP expansion medium until they were 70% confluent and at passage 2 treated with control medium or SMC differentiation medium for 14 days, fixed and then analyzed by immunofluorescence for expression of αSMA, transgelin, calponin and SM-MHC. Primary human bladder smooth muscle cells (HBdSMC) served as the positive control. Nuclei were stained with DAPI. Magnification 20x. Representative of <i>n</i> = 3.</p

    Levels of intracellular Ca<sup>2+</sup>.

    No full text
    <p>Ca<sup>2+</sup> imaging of (A) bladder SMCs and (B) differentiated MSCs (d7). K<sup>+</sup>-induced depolarization increased the intracellular Ca<sup>2+</sup> content (black trace). Depolarization in the presence of 50 μM Cd<sup>2+</sup> prevented the Ca<sup>2+</sup> increase (dashed trace). (C) In undifferentiated MSCs (expanded in GMP expansion medium) no transient increase in cytosolic Ca<sup>2+</sup> was observed in response to K+ induced depolarization. Arrow indicates time point in which 15 mM K<sup>+</sup> was added to the bath solution.</p
    corecore