Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Ocul Pharmacol Ther ; 37(1): 60-71, 2021.
Article in English | MEDLINE | ID: mdl-33449859

ABSTRACT

Purpose: To determine the long-term biocompatibility of HyStem® hydrogel in the rabbit eye for use as a carrier for cell or drug delivery into the ocular space. Methods: HyStem hydrogel formulation solidifies ∼20 min after reconstitution, thus can potentially form a solid deposit after injection in situ. To study the ocular disposition of fluorescein-labeled HyStem, we delivered 50 µL/eye over 1 min into the vitreous space of the rabbit. We used 3 Dutch-Belted and 3 New Zealand-pigmented rabbits, all females, delivered the gel into the right eyes, and injected 50 µL BSS Plus into the left eyes as a control. Retinal morphology was assessed by optical coherence tomography (OCT) and white light fundus photography. Fluorescence fundus photography enabled measurement of the clearance of the labeled hydrogel from the posterior chamber. Visual function was evaluated using flash and flicker electroretinography (ERG) pre- and postinjection and at weekly intervals thereafter for 6 weeks. Retinal immunohistochemistry for microglial inflammatory markers was carried out with antiglial fibrillary acidic protein (GFAP) antibody, isolectin B4 (IB4), and 4',6-diamidino-2-phenylindole (DAPI). Results: The gel was successfully delivered into the vitreous space without the formation of a discrete retinal deposit. Fundus imaging, OCT measurements of retinal thickness, and immunohistochemical data indicated an absence of retinal inflammation, and ERG indicated no impact on retinal function. The half-time of HyStem clearance calculated from the loss of fundus fluorescence was 3.9 days. Conclusions: HyStem hydrogel appears to be biocompatible in the ocular space of a large eye and safe for long-term intraocular application.


Subject(s)
Biocompatible Materials/administration & dosage , Eye/drug effects , Hydrogels/administration & dosage , Animals , Drug Tolerance , Female , Injections, Intraocular , Rabbits
2.
Cancer Immunol Immunother ; 69(9): 1737-1749, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32333082

ABSTRACT

Combination immunotherapy targeting the PD-1 and CTLA-4 checkpoint inhibitor pathways provides substantial clinical benefit in patients with advanced-stage cancer but at the risk of dose-limiting inflammatory and autoimmune toxicity. The delicate balance that exists between unleashing tumor killing and promoting systemic autoimmune toxicity represents a major clinical challenge. We hypothesized that targeting anti-CTLA-4 so that it perfuses tumor-draining lymph nodes would provide a significant therapeutic advantage and developed an injectable hydrogel with controlled antibody release characteristics for this purpose. Injection of hydrogel-encapsulated anti-CTLA-4 at a peri-tumor location (MC-38 tumor model) produced dose-dependent antitumor responses and survival that exceeded those by anti-CTLA-4 alone (p < 0.05). Responses to 100 µg of targeted anti-CTLA-4 also equaled or exceeded those observed with a series of systemic injections delivering 600 µg (p < 0.05). While preserving antitumor activity, this approach resulted in serum anti-CTLA-4 exposure (area under the curve) that averaged only 1/16th of that measured with systemic therapy. Consistent with the marked differences in systemic exposure, systemic anti-CTLA-4 stimulated the onset of autoimmune thyroiditis in iodide-exposed NOD.H-2h4 mice, as measured by anti-thyroglobulin antibody titer, while hydrogel-encapsulated anti-CTLA-4 had a minimal effect (p ≤ 0.01). At the same time, this targeted low-dose anti-CTLA-4 approach synergized well with systemic anti-PD-1 to control tumor growth and resulted in a high frequency of complete responders that were immune to tumor re-challenge at a distant site. We conclude that targeted and controlled delivery of low-dose anti-CTLA-4 has the potential to improve the benefit-risk ratio associated with combination checkpoint inhibitor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , CTLA-4 Antigen/immunology , Delayed-Action Preparations/pharmacology , Immunity/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Autoimmunity/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Drug Synergism , Female , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
3.
Int J Mol Sci ; 19(12)2018 Nov 28.
Article in English | MEDLINE | ID: mdl-30486515

ABSTRACT

Ischemic stroke is a leading cause of death and disability worldwide. Potential therapeutics aimed at neural repair and functional recovery are limited in their blood-brain barrier permeability and may exert systemic or off-target effects. We examined the effects of brain-derived neurotrophic factor (BDNF), delivered via an extended release HyStem®-C hydrogel implant or vehicle, on sensorimotor function, infarct volume, and neuroinflammation, following permanent distal middle cerebral artery occlusion (dMCAo) in rats. Eight days following dMCAo or sham surgery, treatments were implanted directly into the infarction site. Rats received either vehicle, BDNF-only (0.167 µg/µL), hydrogel-only, hydrogel impregnated with 0.057 µg/µL of BDNF (hydrogel + BDNFLOW), or hydrogel impregnated with 0.167 µg/µL of BDNF (hydrogel + BDNFHIGH). The adhesive removal test (ART) and 28-point Neuroscore (28-PN) were used to evaluate sensorimotor function up to two months post-ischemia. The hydrogel + BDNFHIGH group showed significant improvements on the ART six to eight weeks following treatment and their behavioral performance was consistently greater on the 28-PN. Infarct volume was reduced in rats treated with hydrogel + BDNFHIGH as were levels of microglial, phagocyte, and astrocyte marker immunoexpression in the corpus striatum. These data suggest that targeted intracerebral delivery of BDNF using hydrogels may mitigate ischemic brain injury and restore functional deficits by reducing neuroinflammation.


Subject(s)
Brain Ischemia/drug therapy , Brain-Derived Neurotrophic Factor/therapeutic use , Hydrogels/chemistry , Inflammation/drug therapy , Stroke/drug therapy , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Immunohistochemistry , Infarction, Middle Cerebral Artery/drug therapy , Male , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
4.
J Cereb Blood Flow Metab ; 37(3): 1030-1045, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27174996

ABSTRACT

Stroke is the leading cause of adult disability. Systemic delivery of candidate neural repair therapies is limited by the blood-brain barrier and off-target effects. We tested a bioengineering approach for local depot release of BDNF from the infarct cavity for neural repair in chronic periods after stroke. The brain release levels of a hyaluronic acid hydrogel + BDNF were tested in several stroke models in mouse (strains C57Bl/6, DBA) and non-human primate ( Macaca fascicularis) and tracked with MRI. The behavioral recovery effects of hydrogel + BDNF and the effects on tissue repair outcomes were determined. Hydrogel-delivered BDNF diffuses from the stroke cavity into peri-infarct tissue over 3 weeks in two mouse stroke models, compared with 1 week for direct BDNF injection. Hydrogel delivery of BDNF promotes recovery of motor function. Mapping of motor system connections indicates that hydrogel-BDNF induces axonal sprouting within existing cortical and cortico-striatal systems. Pharmacogenetic studies show that hydrogel-BDNF induces the initial migration of immature neurons into the peri-infarct cortex and their long-term survival. In chronic stroke in the non-human primate, hydrogel-released BDNF can be detected up to 2 cm from the infarct, a distance relevant to human functional recovery in stroke. The hydrogel can be tracked by MRI in mouse and primate.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Neurogenesis/drug effects , Recovery of Function/drug effects , Stroke/drug therapy , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Drug Delivery Systems/methods , Macaca fascicularis , Magnetic Resonance Imaging , Mice , Stroke/physiopathology
5.
Acta Biomater ; 43: 122-138, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27475528

ABSTRACT

UNLABELLED: Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVß1 and α5ß1, and several laminin binding integrins (α7ß1, α6ß1, α3ß1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE: Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.


Subject(s)
Blood Vessels/physiology , Fibrin/pharmacology , Hyaluronic Acid/pharmacology , Laminin/pharmacology , Neural Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Blood Vessels/drug effects , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Integrins/metabolism , Neovascularization, Physiologic/drug effects , Neural Stem Cells/drug effects , Polymerization/drug effects , Salmon
6.
Adv Healthc Mater ; 4(11): 1664-74, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26059746

ABSTRACT

Validation of a high-throughput compatible 3D hyaluronic acid hydrogel coculture of cancer cells with stromal cells. The multilayered hyaluronic acid hydrogels improve drug screening predictability as evaluated with a panel of clinically relevant chemotherapeutics in both prostate and endometrial cancer cell lines compared to 2D culture.


Subject(s)
High-Throughput Screening Assays/methods , Hyaluronic Acid/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Automation , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Drug Evaluation, Preclinical , High-Throughput Screening Assays/instrumentation , Humans , Microscopy, Confocal , Stromal Cells/cytology , Stromal Cells/metabolism
7.
Biosensors (Basel) ; 4(1): 28-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24791214

ABSTRACT

Micro-scale printing and patterning of living cells has multiple applications including tissue engineering, cell signaling assays, and the fabrication of cell-based biosensors. In this work, a molecular printing instrument, the Bioforce Nano eNabler, was modified to enable micron-scale -quill-pen based printing of mammalian cells in a 3D hyaluronan/gelatin based hydrogel. Specifically, photo-initiated -thiol-ene click chemistry was used to couple the thiol groups of thiolated hyaluronan/thiolated gelatin to the alkene groups of 4-arm polyethylene glycol (PEG)-norbornene molecules. Rapid photopolymerization enabled direct printing and controlled curing of living cells within the hydrogel matrix. The resulting hydrogels were biocompatible with human adipose-derived stem cells, NIH-3T3 cells, and mouse embryonic stem cells. The utility of this printing approach was also explored for cell-based biosensors. Micro-printed cells expressing a redox sensitive variant of the green fluorescent protein (roGFP-R12) showed a measurable fluorescent response to addition of oxidizing and then reducing agents. This work represents a novel approach to micron-scale cell patterning, and its potential for living, cell-based biosensors.

8.
Acta Biomater ; 10(1): 94-103, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24096152

ABSTRACT

Future ophthalmic therapeutics will require the sustained delivery of bioactive proteins and nucleic acid-based macromolecules and/or provide a suitable microenvironment for the localization and sustenance of reparative progenitor cells after transplantation into or onto the eye. Water-rich hydrogels are ideal vehicles for such cargo, but few have all the qualities desired for novel ophthalmic use, namely in situ gelation speed, cytocompatibility, biocompatibility and capacity to functionalize. We describe here the development of an ophthalmic-compatible crosslinking system using oxidized glutathione (GSSG), a physiologically relevant molecule with a history of safe use in humans. When GSSG is used in conjunction with an existing hyaluronate-based, in situ crosslinkable hydrogel platform, gels form in less than 5 min using the thiol-disulfide exchange reaction. This GSSG hydrogel supports the 3-D culture of adipose-derived stem cells in vitro and shows biocompatibility in preliminary intracutaneous and subconjunctival experiments in vivo. In addition, the thiol-disulfide exchange reaction can also be used in conjunction with other thiol-compatible chemistries to covalently link peptides for more complex formulations. These data suggest that this hydrogel could be well suited for local ocular delivery, focusing initially on front of the eye therapies. Subsequent uses of the hydrogel include delivery of back of the eye treatments and eventually into other soft, hyaluronan-rich tissues such as those from the liver and brain.


Subject(s)
Cross-Linking Reagents/pharmacology , Glutathione Disulfide/pharmacology , Hyaluronic Acid/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Ophthalmology , Sulfhydryl Compounds/pharmacology , Adipose Tissue/cytology , Animals , Eye/drug effects , Glutathione Disulfide/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Injections , Oligopeptides/pharmacology , Rabbits , Skin/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Sulfhydryl Compounds/chemistry
9.
Biomaterials ; 35(1): 71-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120037

ABSTRACT

Changes in tissue and organ stiffness occur during development and are frequently symptoms of disease. Many cell types respond to the stiffness of substrates and neighboring cells in vitro and most cell types increase adherent area on stiffer substrates that are coated with ligands for integrins or cadherins. In vivo cells engage their extracellular matrix (ECM) by multiple mechanosensitive adhesion complexes and other surface receptors that potentially modify the mechanical signals transduced at the cell/ECM interface. Here we show that hyaluronic acid (also called hyaluronan or HA), a soft polymeric glycosaminoglycan matrix component prominent in embryonic tissue and upregulated during multiple pathologic states, augments or overrides mechanical signaling by some classes of integrins to produce a cellular phenotype otherwise observed only on very rigid substrates. The spread morphology of cells on soft HA-fibronectin coated substrates, characterized by formation of large actin bundles resembling stress fibers and large focal adhesions resembles that of cells on rigid substrates, but is activated by different signals and does not require or cause activation of the transcriptional regulator YAP. The fact that HA production is tightly regulated during development and injury and frequently upregulated in cancers characterized by uncontrolled growth and cell movement suggests that the interaction of signaling between HA receptors and specific integrins might be an important element in mechanical control of development and homeostasis.


Subject(s)
Hyaluronic Acid/pharmacology , Integrins/physiology , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , 3T3 Cells , Animals , Cell Proliferation , Cells, Cultured , Extracellular Matrix/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Mice , Microscopy, Atomic Force , Rats , Rats, Sprague-Dawley
10.
Tissue Eng Part A ; 19(1-2): 135-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22953745

ABSTRACT

Control of retinal progenitor cell (RPC) survival, delivery, and differentiation following transplantation into the retina remains a challenge. This is largely due to the use of culture systems that involve poorly defined animal products and do not mimic the natural developmental milieu. We describe the use of hyaluronic acid (HA) based hydrogels to encapsulate mouse RPCs and a delivery system for injectable tissue engineering. We selected HA because of its role in early development and as a feeder layer in stem cell cultures, and the relative ease with which various parameters can be controlled (e.g., hydrogel architecture, mechanics, and degradation). When encapsulated in three-dimensional HA hydrogels, RPCs maintained their undifferentiated state and readily formed neurospheres. These hydrogels were viscous solutions, exhibiting properties ideal for delivery to a subretinal space. The transplants caused very little disruption to the host retinal architecture. Hydrogels were completely degraded and RPCs distributed evenly in the subretinal space by week 3 and expressed the mature photoreceptor marker recoverin. HA hydrogels, with their developmentally relevant composition and malleable physical properties, provide a unique microenvironment for self renewal and differentiation of RPCs for retinal repair.


Subject(s)
Hyaluronic Acid/chemistry , Hydrogels/chemistry , Retina/cytology , Stem Cell Transplantation/methods , Stem Cells/cytology , Tissue Engineering/methods , Animals , Mice
11.
Acta Biomater ; 8(12): 4200-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22776825

ABSTRACT

The current practice of cell therapy, in which multipotent or terminally differentiated cells are injected into tissues or intravenously, is inefficient. Few therapeutic cells are retained at the site of administration and engraftment is low. An injectable and biologically appropriate vehicle for delivery, retention, growth and differentiation of therapeutic cells is needed to improve the efficacy of cell therapy. We focus on a hyaluronan-based semi-synthetic extracellular matrix (sECM), HyStem®, which is a manufacturable, approvable and affordable clinical product. The composition of this sECM can be customized for use with mesenchymal stem cells as well as cells derived from embryonic or induced pluripotent sources. In addition, it can support therapeutic uses of progenitor and mature cell populations obtained from skin, fat, liver, heart, muscle, bone, cartilage, nerves and other tissues. This overview presents four pre-clinical uses of HyStem® for cell therapy to repair injured vocal folds, improve post-myocardial infarct heart function, regenerate damaged liver tissue and restore brain function following ischemic stroke. Finally, we address the real-world limitations - manufacture, regulation, market acceptance and financing - surrounding cell therapy and the development of clinical combination products.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Extracellular Matrix/transplantation , Hyaluronic Acid/therapeutic use , Brain Ischemia/therapy , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Liver Regeneration , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Stroke/therapy , Vocal Cords
12.
Macromol Biosci ; 12(8): 1034-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22730306

ABSTRACT

These studies provide evidence for the ability of a commercially available, defined, hyaluronan-gelatin hydrogel, HyStem-C™, to maintain both mouse embryonic stem cells (mESCs) and human induced pluripotent stem cells (hiPSCs) in culture while retaining their growth and pluripotent characteristics. Growth curve and doubling time analysis show that mESCs and hiPSCs grow at similar rates on HyStem-C™ hydrogels and mouse embryonic fibroblasts and Matrigel™, respectively. Immunocytochemistry, flow cytometry, gene expression and karyotyping reveal that both human and murine pluripotent cells retain a high level of pluripotency on the hydrogels after multiple passages. The addition of fibronectin to HyStem-C™ enabled the attachment of hiPSCs in a xeno-free, fully defined medium.


Subject(s)
Biocompatible Materials/chemistry , Embryonic Stem Cells/cytology , Gelatin/chemistry , Hyaluronic Acid/chemistry , Induced Pluripotent Stem Cells/cytology , Animals , Biocompatible Materials/pharmacology , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibronectins/chemistry , Gelatin/pharmacology , Humans , Hyaluronic Acid/pharmacology , Hydrogels , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Lewis X Antigen/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Tissue Engineering , Tissue Scaffolds
13.
Biomaterials ; 33(21): 5317-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22560668

ABSTRACT

The vast majority of cells delivered into the heart by conventional means are lost within the first 24 h. Methods are needed to enhance cell retention, so as to minimize loss of precious material and maximize effectiveness of the therapy. We tested a cell-hydrogel delivery strategy. Cardiosphere-derived cells (CDCs) were grown from adult human cardiac biopsy specimens. In situ polymerizable hydrogels made of hyaluronan and porcine gelatin (Hystem(®)-C™) were formulated as a liquid at room temperature so as to gel within 20 min at 37 °C. CDC viability and migration were not compromised in Hystem-C™. Myocardial infarction was created in SCID mice and CDCs were injected intramyocardially in the infarct border zone. Real-time PCR revealed engraftment of CDCs delivered in Hystem-C™ was increased by nearly an order of magnitude. LVEF (left ventricular ejection fraction) deteriorated in the control (PBS only) group over the 3-week time course. Hystem-C™ alone or CDCs alone preserved LVEF relative to baseline, while CDCs delivered in Hystem-C™ resulted in a sizable boost in LVEF. Heart morphometry revealed the greatest attenuation of LV remodeling in the CDC + Hystem-C™ group. Histological analysis suggested cardiovascular differentiation of the CDCs in Hystem-C™. However, the majority of functional benefit is likely from paracrine mechanisms such as tissue preservation and neovascularization. A CDC/hydrogel formulation suitable for catheter-based intramyocardial injection exhibits superior engraftment and functional benefits relative to naked CDCs.


Subject(s)
Gelatin/pharmacology , Hyaluronic Acid/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Myocardium/cytology , Polymerization/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/transplantation , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Male , Mice , Microscopy, Fluorescence , Neovascularization, Physiologic/drug effects , Spheroids, Cellular/drug effects , Tissue Preservation
14.
Respir Res ; 12: 8, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21232113

ABSTRACT

BACKGROUND: A newly identified mechanism of smooth muscle relaxation is the interaction between the small heat shock protein 20 (HSP20) and 14-3-3 proteins. Focusing upon this class of interactions, we describe here a novel drug target screening approach for treating airflow obstruction in asthma. METHODS: Using a high-throughput fluorescence polarization (FP) assay, we screened a library of compounds that could act as small molecule modulators of HSP20 signals. We then applied two quantitative, cell-based biophysical methods to assess the functional efficacy of these molecules and rank-ordered their abilities to relax isolated human airway smooth muscle (ASM). Scaling up to the level of an intact tissue, we confirmed in a concentration-responsive manner the potency of the cell-based hit compounds. RESULTS: Among 58,019 compound tested, 268 compounds caused 20% or more reduction of the polarized emission in the FP assay. A small subset of these primary screen hits, belonging to two scaffolds, caused relaxation of isolated ASM cell in vitro and attenuated active force development of intact tissue ex vivo. CONCLUSIONS: This staged biophysical screening paradigm provides proof-of-principle for high-throughput and cost-effective discovery of new small molecule therapeutic agents for obstructive lung diseases.


Subject(s)
14-3-3 Proteins/metabolism , Bronchodilator Agents/pharmacology , Drug Discovery/methods , HSP20 Heat-Shock Proteins/metabolism , Lung Diseases, Obstructive/drug therapy , Lung/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Animals , Bronchodilator Agents/chemistry , Cattle , Dose-Response Relationship, Drug , Fluorescence Polarization , Fourier Analysis , High-Throughput Screening Assays , Humans , In Vitro Techniques , Lung/metabolism , Lung Diseases, Obstructive/metabolism , Magnetics , Male , Molecular Structure , Muscle, Smooth/metabolism , Phosphorylation , Rats , Rats, Inbred F344 , Reproducibility of Results , Signal Transduction/drug effects , Small Molecule Libraries , Structure-Activity Relationship , Surface Plasmon Resonance , Time Factors
15.
Anal Biochem ; 317(2): 255-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12758265

ABSTRACT

High-throughput purification of affinity-tagged fusion proteins is currently one of the fastest developing areas of molecular proteomics. A prerequisite for success in protein purification is sufficient soluble protein expression of the target protein in a heterologous host. Hence, a fast and quantitative evaluation of the soluble-protein levels in an expression system is one of the key steps in the entire process. Here we describe a high-throughput expression screen for affinity-tagged fusion proteins based on an enzyme linked immunofiltration assay (ELIFA). An aliquot of a crude Escherichia coli extract containing the analyte, an affinity-tagged protein, is adsorbed onto the membrane. Subsequent binding of specific antibodies followed by binding of a secondary antibody horseradish peroxidase (HRP) complex then allows quantitative evaluation of the analyte using tetramethylbenzidine as the substrate for HRP. The method is accurate and quantitative, as shown by comparison with results from western blotting and an enzymatic glutathione S-transferase (GST) assay. Furthermore, it is a far more rapid assay and less cumbersome than western blotting, lending itself more readily to high-throughput analysis. It can be used at the expression level (cell lysates) or during the subsequent purification steps to monitor yield of specific protein.


Subject(s)
Chromatography, Affinity/methods , Enzyme-Linked Immunosorbent Assay/methods , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Blotting, Western , Escherichia coli/genetics , Gene Expression , Glutathione Transferase/genetics , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Recombinant Fusion Proteins/genetics , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...