Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Adv Funct Mater ; 34(3)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38707790

ABSTRACT

Skeletal muscle connective tissue (MCT) surrounds myofiber bundles to provide structural support, produce force transduction from tendons, and regulate satellite cell differentiation during muscle regeneration. Engineered muscle tissue composed of myofibers layered within MCT has not yet been developed. Herein, a bioengineering strategy to create MCT-layered myofibers through the development of stem cell fate-controlling biomaterials that achieve both myogenesis and fibroblast differentiation in a locally controlled manner at the single construct is introduced. The reciprocal role of transforming growth factor-beta 1 (TGF-ß1) and its inhibitor as well as 3D matrix stiffness to achieve co-differentiation of MCT fibroblasts and myofibers from a human-induced pluripotent stem cell (hiPSC)-derived paraxial mesoderm is studied. To avoid myogenic inhibition, TGF-ß1 is conjugated on the gelatin-based hydrogel to control the fibroblasts' populations locally; the TGF-ß1 degrades after 2 weeks, resulting in increased MCT-specific extracellular matrix (ECM) production. The locations of myofibers and fibroblasts are precisely controlled by using photolithography and co-axial wet spinning techniques, which results in the formation of MCT-layered functional myofibers in 3D constructs. This advanced engineering strategy is envisioned as a possible method for obtaining biomimetic human muscle grafts for various biomedical applications.

2.
Front Cell Dev Biol ; 11: 1138504, 2023.
Article in English | MEDLINE | ID: mdl-36936695

ABSTRACT

Drug nephrotoxicity is a common healthcare problem in hospitalized patients and a major limitation during drug development. Multi-segmented kidney organoids derived from human pluripotent stem cells may complement traditional cell culture and animal experiments for nephrotoxicity assessment. Here we evaluate the capability of kidney organoids to investigate drug toxicity in vitro. Kidney organoids express renal drug transporters, OAT1, OAT3, and OCT2, while a human proximal tubular cell line shows the absence of OAT1 and OAT3. Tenofovir and aristolochic acid (AA) induce proximal tubular injury in organoids which is ameliorated by an OAT inhibitor, probenecid, without damage to podocytes. Similarly, cisplatin causes proximal tubular damage that can be relieved by an OCT inhibitor, cimetidine, collectively suggesting the presence of functional OATs and OCTs in organoid proximal tubules. Puromycin aminonucleoside (PAN) induced segment-specific injury in glomerular podocytes in kidney organoids in the absence of tubular injury. Reporter organoids were generated with an ATP/ADP biosensor, which may be applicable to high-throughput screening in the future. In conclusion, the kidney organoid is a useful tool for toxicity assessment in the multicellular context and may contribute to nephrotoxicity assessment during drug development.

3.
Biomaterials ; 296: 122058, 2023 05.
Article in English | MEDLINE | ID: mdl-36841214

ABSTRACT

Volumetric muscle loss (VML), which refers to a composite skeletal muscle defect, most commonly heals by scarring and minimal muscle regeneration but substantial fibrosis. Current surgical interventions and physical therapy techniques are limited in restoring muscle function following VML. Novel tissue engineering strategies may offer an option to promote functional muscle recovery. The present study evaluates a colloidal scaffold with hierarchical porosity and controlled mechanical properties for the treatment of VML. In addition, as VML results in an acute decrease in insulin-like growth factor 1 (IGF-1), a myogenic factor, the scaffold was designed to slowly release IGF-1 following implantation. The foam-like scaffold is directly crosslinked onto remnant muscle without the need for suturing. In situ 3D printing of IGF-1-releasing porous muscle scaffold onto VML injuries resulted in robust tissue ingrowth, improved muscle repair, and increased muscle strength in a murine VML model. Histological analysis confirmed regeneration of new muscle in the engineered scaffolds. In addition, the scaffolds significantly reduced fibrosis and increased the expression of neuromuscular junctions in the newly regenerated tissue. Exercise training, when combined with the engineered scaffolds, augmented the treatment outcome in a synergistic fashion. These data suggest highly porous scaffolds and exercise therapy, in combination, may be a treatment option following VML.


Subject(s)
Insulin-Like Growth Factor I , Muscular Diseases , Mice , Animals , Porosity , Regeneration , Muscle, Skeletal/physiology , Muscular Diseases/pathology , Tissue Engineering , Fibrosis , Physical Therapy Modalities , Tissue Scaffolds
4.
J Tissue Eng Regen Med ; 15(7): 634-647, 2021 07.
Article in English | MEDLINE | ID: mdl-33880858

ABSTRACT

Incisional hernia is a common complication of hernia repair despite the development of various synthetic and bio-synthetic repair materials. Poor long-term mechanical strength, leading to high recurrence rates, has limited the use of acellular dermal matrices (ADMs) in ventral hernia repair (VHR). Biologically derived meshes have been an area of increasing interest. Still these materials bring the risk of more aggressive immune response and fibrosis in addition to the mechanical failures suffered by the synthetic materials. Platelet-rich plasma (PRP), a growth-factor-rich autologous blood product, has been shown to improve early neovascularization, tissue deposition, and to decrease the rates of recurrence. Here, we demonstrate that PRP promotes the release of growth factors stromal derived factor (SDF)-1, transforming growth factor-beta, and platelet-derived growth factor in a dose-dependent manner. Additionally, we utilize an aortic ring angiogenesis assay to show that PRP promotes angiogenesis in vitro. A rat model of VHR using StratticeTM ADM demonstrates similar findings in vivo, corresponding with the increased expression of vascular endothelial growth factor and collagen type 1 alpha 1. Finally, we show that the molecular and cellular activity initiated by PRP results in an increased mechanical stiffness of the hernia repair mesh over time. Collectively, these data represent an essential step in demonstrating the utility and the mechanism of platelet-derived plasma in biomaterial-aided wound healing and provide promising preclinical data that suggest such materials may improve surgical outcomes.


Subject(s)
Hernia, Ventral/surgery , Herniorrhaphy , Platelet-Rich Plasma/chemistry , Animals , Biomechanical Phenomena , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cross-Linking Reagents/pharmacology , Dermis/drug effects , Disease Models, Animal , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Microvessels/drug effects , Myofibroblasts/cytology , Myofibroblasts/drug effects , Rats, Inbred Lew , Swine , Wound Healing/drug effects
5.
J Tissue Eng Regen Med ; 15(1): 3-13, 2021 01.
Article in English | MEDLINE | ID: mdl-33197147

ABSTRACT

The recurrence of ventral hernias continues to be a problem faced by surgeons, in spite of efforts toward implementing novel repair techniques and utilizing different materials to promote healing. Cadaveric acellular dermal matrices (Alloderm) have shown some promise in numerous surgical subspecialties, but these meshes still suffer from subsequent failure and necessitation of re-intervention. Here, it is demonstrated that the addition of platelet rich plasma to Alloderm meshes temporally modulates both the innate and cytotoxic inflammatory responses to the implanted material. This results in decreased inflammatory cytokine production at early time points, decreased matrix metalloproteinase expression, and decreased CD8+ T cell infiltration. Collectively, these immune effects result in a healing phenotype that is free from mesh thinning and characterized by increased material stiffness.


Subject(s)
Acellular Dermis , Biocompatible Materials , Collagen , Platelet-Rich Plasma , Rats, Inbred Lew , Surgical Mesh , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Collagen/chemistry , Collagen/immunology , Hernia, Ventral/immunology , Hernia, Ventral/surgery , Male , Platelet-Rich Plasma/chemistry , Platelet-Rich Plasma/immunology , Rats
6.
Small ; 16(36): e1907693, 2020 09.
Article in English | MEDLINE | ID: mdl-32643290

ABSTRACT

Current investigations into hazardous nanoparticles (i.e., nanotoxicology) aim to understand the working mechanisms that drive toxicity. This understanding has been used to predict the biological impact of the nanocarriers as a function of their synthesis, material composition, and physicochemical characteristics. It is particularly critical to characterize the events that immediately follow cell stress resulting from nanoparticle internalization. While reactive oxygen species and activation of autophagy are universally recognized as mechanisms of nanotoxicity, the progression of these phenomena during cell recovery has yet to be comprehensively evaluated. Herein, primary human endothelial cells are exposed to controlled concentrations of polymer-functionalized silica nanoparticles to induce lysosomal damage and achieve cytosolic delivery. In this model, the recovery of cell functions lost following endosomal escape is primarily represented by changes in cell distribution and the subsequent partitioning of particles into dividing cells. Furthermore, multilamellar bodies are found to accumulate around the particles, demonstrating progressive endosomal escape. This work provides a set of biological parameters that can be used to assess cell stress related to nanoparticle exposure and the subsequent recovery of cell processes as a function of endosomal escape.


Subject(s)
Endothelial Cells , Nanoparticles , Polymers , Silicon Dioxide , Cell Line , Endosomes/drug effects , Endosomes/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Models, Biological , Nanoparticles/metabolism , Nanoparticles/toxicity , Polymers/chemistry , Silicon Dioxide/toxicity
7.
Pharmaceutics ; 12(2)2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32024134

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) has been demonstrated to be one of the most vital osteogenic factors for bone augmentation. However, its uncontrolled administration has been associated with catastrophic side effects, which compromised its clinical use. To overcome these limitations, we aimed at developing a safer controlled and sustained release of BMP-2, utilizing poly(lactic-co-glycolic acid)-multistage vector composite microspheres (PLGA-MSV). The loading and release of BMP-2 from PLGA-MSV and its osteogenic potential in vitro and in vivo was evaluated. BMP-2 in vitro release kinetics was assessed by ELISA assay. It was found that PLGA-MSV achieved a longer and sustained release of BMP-2. Cell cytotoxicity and differentiation were evaluated in vitro by MTT and alkaline phosphatase (ALP) activity assays, respectively, with rat mesenchymal stem cells. The MTT results confirmed that PLGA-MSVs were not toxic to cells. ALP test demonstrated that the bioactivity of BMP-2 released from the PLGA-MSV was preserved, as it allowed for the osteogenic differentiation of rat mesenchymal stem cells, in vitro. The biocompatible, biodegradable, and osteogenic PLGA-MSVs system could be an ideal candidate for the safe use of BMP-2 in orthopedic tissue engineering applications.

8.
Sci Rep ; 10(1): 172, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932600

ABSTRACT

Despite recent advances in drug delivery, the targeted treatment of unhealthy cells or tissues continues to remain a priority. In cancer (much like other pathologies), delivery vectors are designed to exploit physical and biological features of unhealthy tissues that are not always homogenous across the disease. In some cases, shifting the target from unhealthy tissues to the whole organ can represent an advantage. Specifically, the natural organ-specific retention of nanotherapeutics following intravenous administration as seen in the lung, liver, and spleen can be strategically exploited to enhance drug delivery. Herein, we outline the development of a cell-based delivery system using macrophages as a delivery vehicle. When loaded with a chemotherapeutic payload (i.e., doxorubicin), these cellular vectors (CELVEC) were shown to provide continued release within the lung. This study provides proof-of-concept evidence of an alternative class of biomimetic delivery vectors that capitalize on cell size to provide therapeutic advantages for pulmonary treatments.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Biomimetics , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Lung/metabolism , Macrophages/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Liberation , Liposomes , Lung/cytology , Male , Mice , Mice, Nude , Tissue Distribution
9.
ACS Biomater Sci Eng ; 6(2): 1112-1123, 2020 02 10.
Article in English | MEDLINE | ID: mdl-33464853

ABSTRACT

Engineering tissue-like scaffolds that can mimic the microstructure, architecture, topology, and mechanical properties of native tissues while offering an excellent environment for cellular growth has remained an unmet need. To address these challenges, multicompartment composite fibers are fabricated. These fibers can be assembled through textile processes to tailor tissue-level mechanical and electrical properties independent of cellular level components. Textile technologies also allow control of the distribution of different cell types and the microstructure of fabricated constructs and the direction of cellular growth within the 3D microenvironment. Here, we engineered composite fibers from biocompatible cores and biologically relevant hydrogel sheaths. The fibers are mechanically robust to being assembled using textile processes and could support adhesion, proliferation, and maturation of cell populations important for the engineering of skeletal muscles. We also demonstrated that the changes in the coating of the multicompartment fibers could potentially enhance myogenesis in vitro.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Cell Proliferation , Hydrogels , Muscle, Skeletal
10.
Adv Healthc Mater ; 8(3): e1801146, 2019 02.
Article in English | MEDLINE | ID: mdl-30609312

ABSTRACT

Cardiac tissue is characterized by being dynamic and contractile, imparting the important role of biomechanical cues in the regulation of normal physiological activity or pathological remodeling. However, the dynamic mechanical tension ability also varies due to extracellular matrix remodeling in fibrosis, accompanied with the phenotypic transition from cardiac fibroblasts (CFs) to myofibroblasts. It is hypothesized that the dynamic mechanical tension ability regulates cardiac phenotypic transition within fibrosis in a strain-mediated manner. In this study, a microdevice that is able to simultaneously and accurately mimic the biomechanical properties of the cardiac physiological and pathological microenvironment is developed. The microdevice can apply cyclic compressions with gradient magnitudes (5-20%) and tunable frequency onto gelatin methacryloyl (GelMA) hydrogels laden with CFs, and also enables the integration of cytokines. The strain-response correlations between mechanical compression and CFs spreading, and proliferation and fibrotic phenotype remolding, are investigated. Results reveal that mechanical compression plays a crucial role in the CFs phenotypic transition, depending on the strain of mechanical load and myofibroblast maturity of CFs encapsulated in GelMA hydrogels. The results provide evidence regarding the strain-response correlation of mechanical stimulation in CFs phenotypic remodeling, which can be used to develop new preventive or therapeutic strategies for cardiac fibrosis.


Subject(s)
Extracellular Matrix , Hydrogels/chemistry , Lab-On-A-Chip Devices , Myocardium , Myofibroblasts , Stress, Mechanical , Animals , Cytokines/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Rats , Rats, Sprague-Dawley
11.
Adv Mater Technol ; 4(9)2019 Sep.
Article in English | MEDLINE | ID: mdl-33072853

ABSTRACT

We report the fabrication of a tubular polydimethylsiloxane (PDMS) platform containing arrays of small pores on the wall for modeling blood vessels in vitro. The thin PDMS tubes are produced following our previously reported templating approach, while the pores are subsequently generated using focused laser ablation. As such, when these perforated PDMS tube are populated with a monolayer of endothelial cells on the interior surfaces and embedded within an extracellular matrix (ECM)-like environment, the endothelial cells can sprout out from the tubes into the surrounding matrix through the open pores. When a pair of perforated PDMS tubes are placed in parallel in the matrix, formation of an interconnected network of microvasculature or larger vessels occurs, which is dependent on the flow dynamics within the PDMS tubes. Moreover, when co-cultured with tumor spheroids, the onset of tumor angiogenesis is observed. Our perforated and endothelialized PDMS tubes are believed to enable convenient vascular modeling in vitro and will likely contribute to improved biological studies as well as therapeutic screening.

12.
ACS Biomater Sci Eng ; 5(6): 2953-2964, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-33405598

ABSTRACT

Tendon injuries are frequent and occur in the elderly, young, and athletic populations. The inadequate number of donors combined with many challenges associated with autografts, allografts, xenografts, and prosthetic devices have added to the value of engineering biological substitutes, which can be implanted to repair the damaged tendons. Electrospun scaffolds have the potential to mimic the native tissue structure along with desired mechanical properties and, thus, have attracted noticeable attention. In order to improve the biological responses of these fibrous structures, we designed and fabricated 3D multilayered composite scaffolds, where an electrospun nanofibrous substrate was coated with a thin layer of cell-laden hydrogel. The whole construct composition was optimized to achieve adequate mechanical and physical properties as well as cell viability and proliferation. Mesenchymal stem cells (MSCs) were differentiated by the addition of bone morphogenetic protein 12 (BMP-12). To mimic the natural function of tendons, the cell-laden scaffolds were mechanically stimulated using a custom-built bioreactor. The synergistic effect of mechanical and biochemical stimulation was observed in terms of enhanced cell viability, proliferation, alignment, and tenogenic differentiation. The results suggested that the proposed constructs can be used for engineering functional tendons.

13.
Small ; : e1703509, 2018 Jul 06.
Article in English | MEDLINE | ID: mdl-29978547

ABSTRACT

Chronic wounds are a major health concern and they affect the lives of more than 25 million people in the United States. They are susceptible to infection and are the leading cause of nontraumatic limb amputations worldwide. The wound environment is dynamic, but their healing rate can be enhanced by administration of therapies at the right time. This approach requires real-time monitoring of the wound environment with on-demand drug delivery in a closed-loop manner. In this paper, a smart and automated flexible wound dressing with temperature and pH sensors integrated onto flexible bandages that monitor wound status in real-time to address this unmet medical need is presented. Moreover, a stimuli-responsive drug releasing system comprising of a hydrogel loaded with thermo-responsive drug carriers and an electronically controlled flexible heater is also integrated into the wound dressing to release the drugs on-demand. The dressing is equipped with a microcontroller to process the data measured by the sensors and to program the drug release protocol for individualized treatment. This flexible smart wound dressing has the potential to significantly impact the treatment of chronic wounds.

14.
Adv Healthc Mater ; 7(11): e1701347, 2018 06.
Article in English | MEDLINE | ID: mdl-29663706

ABSTRACT

Bioprinting has emerged as a promising tool in tissue engineering and regenerative medicine. Various 3D printing strategies have been developed to enable bioprinting of various biopolymers and hydrogels. However, the incorporation of biological factors has not been well explored. As the importance of personalized medicine is becoming more clear, the need for the development of bioinks containing autologous/patient-specific biological factors for tissue engineering applications becomes more evident. Platelet-rich plasma (PRP) is used as a patient-specific source of autologous growth factors that can be easily incorporated to hydrogels and printed into 3D constructs. PRP contains a cocktail of growth factors enhancing angiogenesis, stem cell recruitment, and tissue regeneration. Here, the development of an alginate-based bioink that can be printed and crosslinked upon implantation through exposure to native calcium ions is reported. This platform can be used for the controlled release of PRP-associated growth factors which may ultimately enhance vascularization and stem cell migration.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Precision Medicine/methods , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cell Culture Techniques , Cell Movement/drug effects , Cells, Cultured , Drug Implants/chemistry , Drug Implants/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Hydrogels/chemistry , Ink , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Platelet-Rich Plasma/chemistry
15.
J Tissue Eng Regen Med ; 12(4): 1039-1048, 2018 04.
Article in English | MEDLINE | ID: mdl-29115019

ABSTRACT

Tendons have limited regenerative capacity due to their low cellularity and hypovascular nature, which results in poor clinical outcomes of presently used therapies. As tendon injuries are often observed in active adults, it poses an increasing socio-economic burden on healthcare systems. Currently, suture threads are used during surgical repair to anchor the tissue graft or to connect injured ends. Here, we created composite suture threads coated with a layer of cell-laden hydrogel that can be used for bridging the injured tissue aiming at tendon regeneration. In addition, the fibres can be used to engineer 3-dimensional constructs through textile processes mimicking the architecture and mechanical properties of soft tissues, including tendons and ligaments. Encapsulated human tendon-derived cells migrated within the hydrogel and aligned at the surface of the core thread. An up-regulation of tendon-related genes (scleraxis and tenascin C) and genes involved in matrix remodelling (matrix metalloproteinases 1, matrix metalloproteinases 2) was observed. Cells were able to produce a collagen-rich matrix, remodelling their micro-environment, which is structurally comparable to native tendon tissue.


Subject(s)
Gene Expression Regulation , Hydrogels , Regeneration , Sutures , Tendon Injuries , Tendons/physiology , Humans , Tendon Injuries/metabolism , Tendon Injuries/pathology , Tendon Injuries/therapy
16.
Adv Healthc Mater ; 6(11)2017 Jun.
Article in English | MEDLINE | ID: mdl-28498548

ABSTRACT

Activation of cardiac fibroblasts into myofibroblasts is considered to play an essential role in cardiac remodeling and fibrosis. A limiting factor in studying this process is the spontaneous activation of cardiac fibroblasts when cultured on two-dimensional (2D) culture plates. In this study, a simplified three-dimensional (3D) hydrogel platform of contractile cardiac tissue, stimulated by transforming growth factor-ß1 (TGF-ß1), is presented to recapitulate a fibrogenic microenvironment. It is hypothesized that the quiescent state of cardiac fibroblasts can be maintained by mimicking the mechanical stiffness of native heart tissue. To test this hypothesis, a 3D cell culture model consisting of cardiomyocytes and cardiac fibroblasts encapsulated within a mechanically engineered gelatin methacryloyl hydrogel, is developed. The study shows that cardiac fibroblasts maintain their quiescent phenotype in mechanically tuned hydrogels. Additionally, treatment with a beta-adrenergic agonist increases beating frequency, demonstrating physiologic-like behavior of the heart constructs. Subsequently, quiescent cardiac fibroblasts within the constructs are activated by the exogenous addition of TGF-ß1. The expression of fibrotic protein markers (and the functional changes in mechanical stiffness) in the fibrotic-like tissues are analyzed to validate the model. Overall, this 3D engineered culture model of contractile cardiac tissue enables controlled activation of cardiac fibroblasts, demonstrating the usability of this platform to study fibrotic remodeling.


Subject(s)
Cardiomyopathies/metabolism , Cellular Microenvironment , Fibroblasts/metabolism , Models, Cardiovascular , Myocardium/metabolism , Animals , Cardiomyopathies/pathology , Cells, Cultured , Fibroblasts/pathology , Fibrosis , Hydrogels/chemistry , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
17.
Life Sci ; 171: 60-67, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28039004

ABSTRACT

AIMS: Reestablishment of bladder function in patients with spinal cord injury (SCI) is a clinical priority. Our objectives were to determine whether SCI-localized inhibition of purinergic P2X7 receptors (P2X7R) improve bladder function by decreasing afferent signals mediated by urothelial P2X3R. MAIN METHODS: Systemic inhibition of P2X7R may improve locomotion in rodent SCI models; however, beneficial effects on bladder function and its physiological mechanisms have not been evaluated. We designed a thermosensitive nanohydrogel (NHG) consisting of the P2X7R antagonist brilliant blue-G (BBG) loaded into silica nanoparticles, embedded with poly(d,l-lactic-co-glycolic) acid, and resuspended in 20% pluronic acid. Female Sprague-Dawley rats with a bilateral dorsal lesion at the thoracic T8/T9 region received either 100µl of an empty NHG, or a NHG containing BBG (BBG-NHG) on top of the spinal tissue. Cystometric properties, spinal immunohistochemistry for P2X7R, and bladder immunohistochemistry for P2X3R were evaluated at four weeks post-SCI. KEY FINDINGS: After SCI animals recovered hind-legs use but neurogenic bladder dysfunction remained. SCI rats treated with BBG-NHG for a period of at least two weeks post-SCI experienced fewer non-voiding contractions. The localized inhibition of P2X7R decreased microglia activation. At the lower urinary tract level we observed, unexpectedly, a concomitant reduction of urothelial P2X3 receptors, which are involved in initiation of bladder afferent transmission to start micturition. SIGNIFICANCE: Localized inhibition of P2X7R for two weeks can be associated with reduced number of microglia and attenuated bladder hyperexcitability mediated by downregulation of urothelial P2X3R in rats with neurogenic bladder dysfunction and independently of locomotor improvements.


Subject(s)
Purinergic Antagonists/pharmacology , Receptors, Purinergic P2X3/metabolism , Receptors, Purinergic P2X7/drug effects , Spinal Cord Injuries/metabolism , Urinary Bladder, Neurogenic/drug therapy , Actins/metabolism , Animals , Female , Hydrogels , Purinergic Antagonists/therapeutic use , Rats , Rats, Sprague-Dawley , Urinary Bladder/drug effects , Urinary Bladder/metabolism
18.
J Nanosci Nanotechnol ; 17(2): 977-82, 2017 Feb.
Article in English | MEDLINE | ID: mdl-29671486

ABSTRACT

Solid lipid nanoparticles carrying a chemotherapeutic payload (i.e., temozolomide, TMZ) were synthesized using ghee, a clarified butter commonly used in traditional medicine and food products. Ghee solid lipid nanoparticles (GSLN) were characterized through dynamic light scattering, scanning electron microscopy, and UV-visible spectrometry. Formulations were generated with varying ratios of surfactant to lipid, resulting in a maximum TMZ entrapment efficiency of ˜70%. Optimal formulations were found to have an average size and polydispersity of ˜220 nm and 0.340, respectively. Release kinetics revealed TMZ-loaded GSLN (TMZ@GSLN) retained 10% of its pay-load at 2 h with ˜53% released in 5 h. Metabolic activity on human umbilical vein endothelial cells (HUVEC) revealed GSLN treatment resulted in an increase in viability following 3 d while treatment of glioblastoma LN-229 cells with TMZ@GSLN resulted in a significant decrease. Evaluation of diffusion of TMZ across a reconstructed HUVEC monolayer demonstrated TMZ@GSLN resulted in a significantly higher diffusion of drug when compared to free TMZ. This data suggests GSLN pose a promising delivery vehicle for TMZ-based therapeutics. Collectively, this data demonstrates GSLN exhibit favorable drug carrier properties with anti-proliferative properties in glioblastoma cancer cells.


Subject(s)
Drug Carriers , Ghee , Nanoparticles/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/chemistry , Dacarbazine/pharmacokinetics , Dacarbazine/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Human Umbilical Vein Endothelial Cells , Humans , Temozolomide
19.
Appl Microbiol Biotechnol ; 101(2): 475-491, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28013407

ABSTRACT

In the post-genome age, proteomics is receiving significant attention because they provide an invaluable source of biological structures and functions at the protein level. The search for disease-specific biomarkers for diagnostic and/or therapeutic applications is one of the areas that proteomics is having a significant impact. Thus, the identification of a "good" biomarker enables a more accurate early diagnosis and prognosis of disease. Rapid advancements in mass spectrometry (MS) instrumentation, liquid chromatography MS (LCMS), protein microarray technology, and other protein profiling methodologies have a substantial expansion of our toolbox to identify disease-specific protein and peptide biomarkers. This review covers a selection of widely used proteomic technologies for biomarker discovery. In addition, we describe the most commonly used approaches for diagnosis based on proteomic biomarkers and further discuss trends and critical challenges during development of cost-effective rapid diagnostic tests and microfluidic diagnostic systems based on proteomic biomarkers.


Subject(s)
Biomarkers/analysis , Diagnostic Tests, Routine/methods , Proteomics/methods
20.
J Biomed Mater Res B Appl Biomater ; 105(8): 2206-2217, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27438342

ABSTRACT

Current treatments in hypoplastic left heart syndrome (HLHS) include multiple surgeries to refunctionalize the right ventricle and/or transplant. The development of a tissue-engineered left ventricle (LV) would provide a therapeutic option to overcome the inefficiencies and limitations associated with current treatment options. This study provides a foundation for the development and fabrication of the bioengineered open ventricle (BEOV) model. BEOV molds were developed to emulate the human LV geometry; molds were used to produce chitosan scaffolds. BEOV were fabricated by culturing 30 million rat neonatal cardiac cells on the chitosan scaffold. The model demonstrated 57% cell retention following 4days culture. The average biopotential output for the model was 1615 µV. Histological assessment displayed the presence of localized cell clusters, with intercellular and cell-scaffold interactions. The BEOV provides a novel foundation for the development of a 3D bioengineered LV for application in HLHS. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2206-2217, 2017.


Subject(s)
Chitosan/chemistry , Heart Ventricles , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...