18 research outputs found

    Hypoxia Augments Outgrowth Endothelial Cell (OEC) Sprouting and Directed Migration in Response to Sphingosine-1-Phosphate (S1P)

    Get PDF
    Therapeutic angiogenesis provides a promising approach to treat ischemic cardiovascular diseases through the delivery of proangiogenic cells and/or molecules. Outgrowth endothelial cells (OECs) are vascular progenitor cells that are especially suited for therapeutic strategies given their ease of noninvasive isolation from umbilical cord or adult peripheral blood and their potent ability to enhance tissue neovascularization. These cells are recruited to sites of vascular injury or tissue ischemia and directly incorporate within native vascular endothelium to participate in neovessel formation. A better understanding of how OEC activity may be boosted under hypoxia with external stimulation by proangiogenic molecules remains a challenge to improving their therapeutic potential. While vascular endothelial growth factor (VEGF) is widely established as a critical factor for initiating angiogenesis, sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, has recently gained great enthusiasm as a potential mediator in neovascularization strategies. This study tests the hypothesis that hypoxia and the presence of VEGF impact the angiogenic response of OECs to S1P stimulation in vitro. We found that hypoxia altered the dynamically regulated S1P receptor 1 (S1PR1) expression on OECs in the presence of S1P (1.0 mu M) and/or VEGF (1.3 nM). the combined stimuli of S1P and VEGF together promoted OEC angiogenic activity as assessed by proliferation, wound healing, 3D sprouting, and directed migration under both normoxia and hypoxia. Hypoxia substantially augmented the response to S1P alone, resulting in similar to 6.5-fold and similar to 25-fold increases in sprouting and directed migration, respectively. Overall, this report highlights the importance of establishing hypoxic conditions in vitro when studying ischemia-related angiogenic strategies employing vascular progenitor cells.University of California, DavisAmerican Heart Association (AHA)Univ Calif Davis, Dept Biomed Engn, Davis, CA 95616 USAUniversidade Federal de São Paulo, Dept Biophys, São Paulo, BrazilUniv Calif Davis, Dept Neurobiol Physiol & Behav, Davis, CA 95616 USAUniversidade Federal de São Paulo, Dept Biophys, São Paulo, BrazilAmerican Heart Association (AHA): 15PRE22930044Web of Scienc

    PRP and BMAC for Musculoskeletal Conditions via Biomaterial Carriers

    No full text
    Platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMAC) are orthobiologic therapies considered as an alternative to the current therapies for muscle, bone and cartilage. Different formulations of biomaterials have been used as carriers for PRP and BMAC in order to increase regenerative processes. The most common biomaterials utilized in conjunction with PRP and BMAC clinical trials are organic scaffolds and natural or synthetic polymers. This review will cover the combinatorial strategies of biomaterial carriers with PRP and BMAC for musculoskeletal conditions (MsCs) repair and regeneration in clinical trials. The main objective is to review the therapeutic use of PRP and BMAC as a treatment option for muscle, bone and cartilage injuries

    Microgels produced using microfluidic on-chip polymer blending for controlled released of VEGF encoding lentivectors

    No full text
    Alginate hydrogels are widely used as delivery vehicles due to their ability to encapsulate and release a wide range of cargos in a gentle and biocompatible manner. The release of encapsulated therapeutic cargos can be promoted or stunted by adjusting the hydrogel physiochemical properties. However, the release from such systems is often skewed towards burst-release or lengthy retention. To address this, we hypothesized that the overall magnitude of burst release could be adjusted by combining microgels with distinct properties and release behavior. Microgel suspensions were generated using a process we have termed on-chip polymer blending to yield composite suspensions of a range of microgel formulations. In this manner, we studied how alginate percentage and degradation relate to the release of lentivectors. Whereas changes in alginate percentage had a minimal impact on lentivector release, microgel degradation led to a 3-fold increase, and near complete release, over 10 days. Furthermore, by controlling the amount of degradable alginate present within microgels the relative rate of release can be adjusted. A degradable formulation of microgels was used to deliver vascular endothelial growth factor (VEGF)-encoding lentivectors in the chick chorioallantoic membrane (CAM) assay and yielded a proangiogenic response in comparison to the same lentivectors delivered in suspension. The utility of blended microgel suspensions may provide an especially appealing platform for the delivery of lentivectors or similarly sized therapeutics.status: publishe

    Hydrogel biophysical properties instruct coculture-mediated osteogenic potential

    No full text
    Cell-based approaches for bone formation require instructional cues from the surrounding environment. As an alternative to pharmacological strategies or transplanting single cell populations, one approach is to coimplant populations that can establish a new vasculature and differentiate to bone-forming osteoblasts. Mesenchymal stem/stromal cells (MSCs) possess osteogenic potential and produce numerous angiogenic growth factors. Endothelial colony-forming cells (ECFCs) are a subpopulation of endothelial progenitor cells capable of vasculogenesis in vivo and may provide endogenous cues to support MSC function. We investigated the contribution of the carrier biophysical properties to instruct entrapped human MSCs and ECFCs to simultaneously promote their osteogenic and proangiogenic potential. Compared with gels containing MSCs alone, fibrin gels engineered with increased compressive stiffness simultaneously increased the osteogenic and proangiogenic potential of entrapped cocultured cells. ECFCs produced bone morphogenetic protein-2 (BMP-2), a potent osteoinductive molecule, and increases in BMP-2 secretion correlated with gel stiffness. Coculture of MSCs with ECFCs transduced to knockdown BMP-2 production abrogated the osteogenic response to levels observed with MSCs alone. These results demonstrate that physical properties of engineered hydrogels modulate the function of cocultured cells in the absence of inductive cues, thus increasing the translational potential of coimplantation to speed bone formation and repair.U.S. National Institutes of HealthNational Institute of Dental and Craniofacial Research [R03-DE021704]AO Foundation (Davos, Switzerland) [C10-39L]American Heart Association Western States Affiliate Pre-doctoral FellowshipUniv Calif Davis, Dept Biomed Engn, Davis, CA 95616 USAUniv Calif Davis, Sch Med, Dept Orthopaed Surg, Davis, CA 95616 USAUniv Fed Sao Paulo, Dept Biophys, Sao Paulo, BrazilUniv Fed Sao Paulo, Dept Biophys, Sao Paulo, BrazilU.S. National Institutes of Health, National Institute of Dental and Craniofacial Research Grant R03-DE021704, and the AO Foundation (Davos, Switzerland) (C10-39L to J.K.L.). K.M. was supported by the American Heart Association Western States Affiliate Pre-doctoral FellowshipWeb of Scienc

    Injectable alginate hydrogel for enhanced spatiotemporal control of lentivector delivery in murine skeletal muscle

    No full text
    Hydrogels are an especially appealing class of biomaterials for gene delivery vehicles as they can be introduced into the body with minimally invasive procedures and are often applied in tissue engineering and regenerative medicine strategies. In this study, we show for the first time the use of an injectable alginate hydrogel for controlled delivery of lentivectors in the skeletal muscle of murine hindlimb. We propose to alter the release rates of lentivectors through manipulation of the molecular weight distribution of alginate hydrogels. The release of lentivector was tested using two different ratios of low and high molecular weight (MW) alginate polymers (75/25 and 25/75 low/high MW). The interdependency of lentivector release rate and alginate degradation rate was assessed in vitro. Lentivector-loaded hydrogels maintained transduction potential for up to one week in vitro as demonstrated by the continual transduction of HEK-293T cells. Injection of lentivector-loaded hydrogel in vivo led to a sustained level of transgene expression for more than two months while minimizing the copies of lentivirus genome inserted into the genome of murine skeletal muscle cells. This strategy of spatiotemporal control of lentivector delivery from alginate hydrogels may provide a versatile tool to combine gene therapy and biomaterials for applications in regenerative medicine. (C) 2016 Elsevier B.V. All rights reserved.University of California, DavisFAPESP-Sao Paulo, BrazilAmerican Heart AssociationHellman FamilyFAPESP scholarshipHHMI Integrating Medicine into Basic Science fellowshipUniv Fed Sao Paulo, Dept Biophys, Sao Paulo, BrazilUniv Calif Davis, Dept Biomed Engn, 1 Shields Ave, Davis, CA 95616 USAIrmandade Santa Casa Misericordia Sao Paulo, Dept Internal Med, Sao Paulo, BrazilUniv Fed Sao Paulo, Dept Biophys, Sao Paulo, BrazilFAPESP: 2015/20206-8AHA: 15BGIA25730057FAPESP scholarship: 2012/00,753-6Web of Scienc

    Hypoxic culture led to enhanced directed migration towards a gradient of S1P <i>in vitro</i>.

    No full text
    <p>A modified transwell assay was used to quantify directed migration/3D matrix invasion of OECs towards S1P in a fibrin gel (A). Gradients of S1P and VEGF were established by sustained delivery from an alginate hydrogel (B). While combined release of S1P and VEGF resulted in greater migration after 48h in both normoxia and hypoxia, S1P alone was equally sufficient under hypoxia (C). Data are mean ± SD (n = 4). An asterisk indicates statistically significant differences (P<0.05) between conditions in normoxia or hypoxia respectively and N.S. displays no statistically significant difference between conditions.</p

    The combination of S1P and VEGF induced greater OEC proliferation than either individual stimulus under both normoxia and hypoxia.

    No full text
    <p>Data are mean ± SD (n = 6) and normalized to the initial cell seeding number (indicated by dashed line). An asterisk indicates statistically significant differences (P<0.05) between conditions in normoxia or hypoxia respectively.</p
    corecore