Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Biomater ; 167: 425-435, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37321528

ABSTRACT

Regenerative therapeutics for treating peripheral arterial disease are an appealing strategy for creating more durable solutions for limb ischemia. In this work, we performed preclinical testing of an injectable formulation of syndecan-4 proteoliposomes combined with growth factors as treatment for peripheral ischemia delivered in an alginate hydrogel. We tested this therapy in an advanced model of hindlimb ischemia in rabbits with diabetes and hyperlipidemia. Our studies demonstrate enhancement in vascularity and new blood vessel growth with treatment with syndecan-4 proteoliposomes in combination with FGF-2 or FGF-2/PDGF-BB. The effects of the treatments were particularly effective in enhancing vascularity in the lower limb with a 2-4 increase in blood vessels in the treatment group in comparison to the control group. In addition, we demonstrate that the syndecan-4 proteoliposomes have stability for at least 28 days when stored at 4°C to allow transport and use in the hospital environment. In addition, we performed toxicity studies in the mice and found no toxic effects even when injected at high concentration. Overall, our studies support that syndecan-4 proteoliposomes markedly enhance the therapeutic potential of growth factors in the context of disease and may be promising therapeutics for inducing vascular regeneration in peripheral ischemia. STATEMENT OF SIGNIFICANCE: Peripheral ischemia is a common condition in which there is a lack of blood flow to the lower limbs. This condition can lead to pain while walking and, in severe cases, critical limb ischemia and limb loss. In this study, we demonstrate the safety and efficacy of a novel injectable therapy for enhancing revascularization in peripheral ischemia using an advanced large animal model of peripheral vascular disease using rabbits with hyperlipidemia and diabetes.


Subject(s)
Hyperlipidemias , Peripheral Vascular Diseases , Rabbits , Mice , Animals , Syndecan-4/pharmacology , Syndecan-4/therapeutic use , Fibroblast Growth Factor 2 , Neovascularization, Physiologic , Ischemia/therapy , Hindlimb/blood supply , Disease Models, Animal
2.
Biomaterials ; 275: 120924, 2021 08.
Article in English | MEDLINE | ID: mdl-34147716

ABSTRACT

One in 190 Americans is currently living with the loss of a limb resulted from injury, amputation, or neurodegenerative disease. Advanced neuroprosthetic devices combine peripheral neural interfaces with sophisticated prosthetics and hold great potential for the rehabilitation of impaired motor and sensory functions. While robotic prosthetics have advanced very rapidly, peripheral neural interfaces have long been limited by the capability of interfacing with the peripheral nervous system. In this work, we developed a hyperflexible regenerative sieve electrode to serve as a peripheral neural interface. We examined tissue neurovascular integration through this novel device. We demonstrated that we could enhance the neurovascular invasion through the device with directional growth factor delivery. Furthermore, we demonstrated that we could reduce the tissue reaction to the device often seen in peripheral neural interfaces. Finally, we show that we can create a stable tissue device interface in a long-term implantation that does not impede the normal regenerative processes of the nerve. Our study developed an optimal platform for the continued development of hyperflexible sieve electrode peripheral neural interfaces.


Subject(s)
Artificial Limbs , Neurodegenerative Diseases , Electrodes, Implanted , Humans , Nerve Regeneration , Peripheral Nerves
3.
Sci Rep ; 11(1): 9838, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972619

ABSTRACT

Physical activity has been consistently linked to decreased incidence of breast cancer and a substantial increase in the length of survival of patients with breast cancer. However, the understanding of how applied physical forces directly regulate breast cancer remains limited. We investigated the role of mechanical forces in altering the chemoresistance, proliferation and metastasis of breast cancer cells. We found that applied mechanical tension can dramatically alter gene expression in breast cancer cells, leading to decreased proliferation, increased resistance to chemotherapeutic treatment and enhanced adhesion to inflamed endothelial cells and collagen I under fluidic shear stress. A mechanistic analysis of the pathways involved in these effects supported a complex signaling network that included Abl1, Lck, Jak2 and PI3K to regulate pro-survival signaling and enhancement of adhesion under flow. Studies using mouse xenograft models demonstrated reduced proliferation of breast cancer cells with orthotopic implantation and increased metastasis to the skull when the cancer cells were treated with mechanical load. Using high throughput mechanobiological screens we identified pathways that could be targeted to reduce the effects of load on metastasis and found that the effects of mechanical load on bone colonization could be reduced through treatment with a PI3Kγ inhibitor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Breast/pathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Stress, Mechanical , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomechanical Phenomena , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , X-Ray Microtomography , Xenograft Model Antitumor Assays
4.
Nat Biomed Eng ; 5(1): 89-102, 2021 01.
Article in English | MEDLINE | ID: mdl-33483713

ABSTRACT

Using endogenous mesenchymal stem cells for treating myocardial infarction and other cardiovascular conditions typically results in poor efficacy, in part owing to the heterogeneity of the harvested cells and of the patient responses. Here, by means of high-throughput screening of the combinatorial space of mechanical-strain level and of the presence of particular kinase inhibitors, we show that human mesenchymal stem cells can be mechanically and pharmacologically conditioned to enhance vascular regeneration in vivo. Mesenchymal stem cells conditioned to increase the activation of signalling pathways mediated by Smad2/3 (mothers against decapentaplegic homolog 2/3) and YAP (Yes-associated protein) expressed markers that are associated with pericytes and endothelial cells, displayed increased angiogenic activity in vitro, and enhanced the formation of vasculature in mice after subcutaneous implantation and after implantation in ischaemic hindlimbs. These effects were mediated by the crosstalk of endothelial-growth-factor receptors, transforming-growth-factor-beta receptor type 1 and vascular-endothelial-growth-factor receptor 2. Mechanical and pharmacological conditioning can significantly enhance the regenerative properties of mesenchymal stem cells.


Subject(s)
Biomechanical Phenomena/physiology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic/physiology , Regeneration/physiology , Adult , Animals , Female , Humans , Ischemia , Male , Mesenchymal Stem Cell Transplantation , Mice , Neovascularization, Physiologic/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Growth Factor/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Young Adult
5.
Biotechniques ; 69(6): 443-449, 2020 12.
Article in English | MEDLINE | ID: mdl-33108897

ABSTRACT

The quantitative analysis of blood vessel networks is an important component in many animal models of disease. We describe a nondestructive technique for blood vessel imaging that visualizes in situ vasculature in harvested tissues. The method allows for further analysis of the same tissues with histology and other methods that can be performed on fixed tissue. Consequently, it can easily be incorporated upstream to analysis methods to augment these with a three-dimensional reconstruction of the vascular network in the tissues to be analyzed. The method combines iodine-enhanced micro-computed tomography with a deep learning algorithm to segment vasculature within tissues. The procedure is relatively simple and can provide insight into complex changes in the vascular structure in the tissues.


Subject(s)
Blood Vessels/diagnostic imaging , Imaging, Three-Dimensional/methods , Animals , Brain/blood supply , Brain/diagnostic imaging , Heart/diagnostic imaging , Iodine/chemistry , Male , Neural Networks, Computer , Peripheral Nerves/blood supply , Peripheral Nerves/diagnostic imaging , Rats, Sprague-Dawley , Staining and Labeling , X-Ray Microtomography
6.
Integr Biol (Camb) ; 12(1): 1-11, 2020 02 22.
Article in English | MEDLINE | ID: mdl-31965190

ABSTRACT

Tumor-initiating cells (TICs), a subpopulation of cancerous cells with high tumorigenic potential and stem-cell-like properties, drive tumor progression and are resistant to conventional therapies. Identification and isolation of TICs are limited by their low frequency and lack of robust markers. Here, we characterize the heterogeneous adhesive properties of a panel of human and murine cancer cells and demonstrate differences in adhesion strength among cells, which exhibit TIC properties and those that do not. These differences in adhesion strength were exploited to rapidly (~10 min) and efficiently isolate cancerous cells with increased tumorigenic potential in a label-free manner by use of a microfluidic technology. Isolated murine and human cancer cells gave rise to larger tumors with increased growth rate and higher frequency in both immunocompetent and immunocompromised mice, respectively. This rapid and label-free TIC isolation technology has the potential to be a valuable tool for facilitating research into TIC biology and the development of more efficient diagnostics and cancer therapies.


Subject(s)
Carcinogenesis/pathology , Cell Adhesion , Cell Separation/methods , Hydrodynamics , Neoplasms/physiopathology , Neoplastic Stem Cells/pathology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Green Fluorescent Proteins/metabolism , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, SCID , Microfluidics , Signal Transduction , Stress, Mechanical
7.
Adv Drug Deliv Rev ; 146: 97-125, 2019 06.
Article in English | MEDLINE | ID: mdl-30267742

ABSTRACT

The enhancement of wound healing has been a goal of medical practitioners for thousands of years. The development of chronic, non-healing wounds is a persistent medical problem that drives patient morbidity and increases healthcare costs. A key aspect of many non-healing wounds is the reduced presence of vessel growth through the process of angiogenesis. This review surveys the creation of new treatments for healing cutaneous wounds through therapeutic angiogenesis. In particular, we discuss the challenges and advancement that have been made in delivering biologic, pharmaceutical and cell-based therapies as enhancers of wound vascularity and healing.


Subject(s)
Cell- and Tissue-Based Therapy , Neovascularization, Pathologic/therapy , Wound Healing , Animals , Drug Delivery Systems , Humans , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...