Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
SLAS Discov ; 25(2): 163-175, 2020 02.
Article in English | MEDLINE | ID: mdl-31875412

ABSTRACT

Malfunctions in the basic epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling are implicated in a number of cancers and immunological and neurodegenerative conditions. Within GlaxoSmithKline (GSK) we have utilized a number of variations of the NanoBRET technology for the direct measurement of compound-target engagement within native cellular environments to drive high-throughput, routine structure-activity relationship (SAR) profiling across differing epigenetic targets. NanoBRET is a variation of the bioluminescence resonance energy transfer (BRET) methodology utilizing proteins of interest fused to either NanoLuc, a small, high-emission-intensity luciferase, or HaloTag, a modified dehalogenase enzyme that can be selectively labeled with a fluorophore. The combination of these two technologies has enabled the application of NanoBRET to biological systems such as epigenetic protein-protein interactions, which have previously been challenging. By synergizing target engagement assays with more complex primary cell phenotypic assays, we have been able to demonstrate compound-target selectivity profiles to enhance cellular potency and offset potential liability risks. Additionally, we have shown that in the absence of a robust, cell phenotypic assay, it is possible to utilize NanoBRET target engagement assays to aid chemistry in progressing at a higher scale than would have otherwise been achievable. The NanoBRET target engagement assays utilized have further shown an excellent correlation with more reductionist biochemical and biophysical assay systems, clearly demonstrating the possibility of using such assay systems at scale, in tandem with, or in preference to, lower-throughput cell phenotypic approaches.


Subject(s)
Biological Assay , Epigenesis, Genetic/genetics , Structure-Activity Relationship , Chromatin Assembly and Disassembly/genetics , DNA Methylation/genetics , Fluorescence Resonance Energy Transfer , Histone Code/genetics , Humans , Luciferases/chemistry
2.
Adv Sci (Weinh) ; 5(7): 1700540, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30027026

ABSTRACT

CRISPR/Cas9 technology enables targeted gene editing; yet, the efficiency and specificity remain unsatisfactory, particularly for the nonvirally delivered, plasmid-based CRISPR/Cas9 system. To tackle this, a self-assembled micelle is developed and evaluated for human papillomavirus (HPV) E7 oncogene disruption. The optimized micelle enables effective delivery of Cas9 plasmid with a transient transgene expression profile, benefiting the specificity of Cas9 recognition. Furthermore, the feasibility of using the micelle is explored for another nucleic acid-guided nuclease system, Natronobacterium gregoryi Argonaute (NgAgo). Both systems are tested in vitro and in vivo to evaluate their therapeutic potential. Cas9-mediated E7 knockout leads to significant inhibition of HPV-induced cancerous activity both in vitro and in vivo, while NgAgo does not show significant E7 inhibition on the xenograft mouse model. Collectively, this micelle represents an efficient delivery system for nonviral gene editing, adding to the armamentarium of gene editing tools to advance safe and effective precision medicine-based therapeutics.

3.
Proc Natl Acad Sci U S A ; 115(19): 4903-4908, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29686087

ABSTRACT

Effective and safe delivery of the CRISPR/Cas9 gene-editing elements remains a challenge. Here we report the development of PEGylated nanoparticles (named P-HNPs) based on the cationic α-helical polypeptide poly(γ-4-((2-(piperidin-1-yl)ethyl)aminomethyl)benzyl-l-glutamate) for the delivery of Cas9 expression plasmid and sgRNA to various cell types and gene-editing scenarios. The cell-penetrating α-helical polypeptide enhanced cellular uptake and promoted escape of pCas9 and/or sgRNA from the endosome and transport into the nucleus. The colloidally stable P-HNPs achieved a Cas9 transfection efficiency up to 60% and sgRNA uptake efficiency of 67.4%, representing an improvement over existing polycation-based gene delivery systems. After performing single or multiplex gene editing with an efficiency up to 47.3% in vitro, we demonstrated that P-HNPs delivering Cas9 plasmid/sgRNA targeting the polo-like kinase 1 (Plk1) gene achieved 35% gene deletion in HeLa tumor tissue to reduce the Plk1 protein level by 66.7%, thereby suppressing the tumor growth by >71% and prolonging the animal survival rate to 60% within 60 days. Capable of delivering Cas9 plasmids to various cell types to achieve multiplex gene knock-out, gene knock-in, and gene activation in vitro and in vivo, the P-HNP system offers a versatile gene-editing platform for biological research and therapeutic applications.


Subject(s)
CRISPR-Cas Systems , Cell-Penetrating Peptides , Gene Editing/methods , Gene Transfer Techniques , Nanoparticles/chemistry , Plasmids , Animals , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Mice , NIH 3T3 Cells , Plasmids/chemistry , Plasmids/genetics , Plasmids/pharmacology
4.
Integr Biol (Camb) ; 9(12): 956-967, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29168874

ABSTRACT

In vitro models of the neuromuscular junction (NMJ) are emerging as a valuable tool to study synaptogenesis, synaptic maintenance, and pathogenesis of neurodegenerative diseases. Many models have previously been developed using a variety of cell sources for skeletal muscle and motoneurons. These models can advanced by integrating beneficial features of the native developmental milieu of the NMJ. We created a functional in vitro model of NMJ by bioreactor cultivation of transdifferentiated myocytes and stem cell-derived motoneurons, in the presence of electrical stimulation. In conjunction with a coculture medium, electrical stimulation resulted in improved maturation and function of motoneurons and myocytes, as evidenced by mature cellular structures, increased expression of neuronal and muscular genes, clusterization of acetylcholine receptors (AChRs) in the vicinity of motoneurons, and the response to glutamate stimulation. To validate the model and demonstrate its utility for pharmacological testing, we documented the potency of drugs that affect key pathways during NMJ signal transduction: (i) acetylcholine (ACh) synthesis, (ii) ACh vesicular storage, (iii) ACh synaptic release, (iv) AChR activation, and (v) ACh inactivation in the synaptic cleft. The model properly responded to the drugs in a concentration-dependent manner. We thus propose that this in vitro model of NMJ could be used as a platform in pharmacological screening and controlled studies of neuromuscular diseases.


Subject(s)
Bioreactors , Motor Neurons/drug effects , Muscle Cells/drug effects , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Animals , Cell Separation , Coculture Techniques , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Electric Stimulation , Glutamic Acid/chemistry , Green Fluorescent Proteins/metabolism , Magnetics , Mice , Motor Neurons/physiology , Muscle Cells/cytology , Stem Cells/cytology
5.
Chem Rev ; 117(15): 9874-9906, 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28640612

ABSTRACT

Genome editing offers promising solutions to genetic disorders by editing DNA sequences or modulating gene expression. The clustered regularly interspaced short palindromic repeats (CRISPR)/associated protein 9 (CRISPR/Cas9) technology can be used to edit single or multiple genes in a wide variety of cell types and organisms in vitro and in vivo. Herein, we review the rapidly developing CRISPR/Cas9-based technologies for disease modeling and gene correction and recent progress toward Cas9/guide RNA (gRNA) delivery based on viral and nonviral vectors. We discuss the relative merits of delivering the genome editing elements in the form of DNA, mRNA, or protein, and the opportunities of combining viral delivery of a transgene encoding Cas9 with nonviral delivery of gRNA. We highlight the lessons learned from nonviral gene delivery in the past three decades and consider their applicability for CRISPR/Cas9 delivery. We also include a discussion of bioinformatics tools for gRNA design and chemical modifications of gRNA. Finally, we consider the extracellular and intracellular barriers to nonviral CRISPR/Cas9 delivery and propose strategies that may overcome these barriers to realize the clinical potential of CRISPR/Cas9-based genome editing.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Gene Transfer Techniques , Genetic Therapy/methods , Models, Biological , Humans
6.
Sci Rep ; 7: 40285, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071742

ABSTRACT

Transdifferentiation has been described as a novel method for converting human fibroblasts into induced cardiomyocyte-like cells. Such an approach can produce differentiated cells to study physiology or pathophysiology, examine drug interactions or toxicities, and engineer cardiac tissues. Here we describe the transdifferentiation of human dermal fibroblasts towards the cardiac cell lineage via the induced expression of transcription factors GATA4, TBX5, MEF2C, MYOCD, NKX2-5, and delivery of microRNAs miR-1 and miR-133a. Cells undergoing transdifferentiation expressed ACTN2 and TNNT2 and partially organized their cytoskeleton in a cross-striated manner. The conversion process was associated with significant upregulation of a cohort of cardiac-specific genes, activation of pathways associated with muscle contraction and physiology, and downregulation of fibroblastic markers. We used a genetically encoded calcium indicator and readily detected active calcium transients although no spontaneous contractions were observed in transdifferentiated cells. Finally, we determined that inhibition of Janus kinase 1, inhibition of Glycogen synthase kinase 3, or addition of NRG1 significantly enhanced the efficiency of transdifferentiation. Overall, we describe a method for achieving transdifferentiation of human dermal fibroblasts into induced cardiomyocyte-like cells via transcription factor overexpression, microRNA delivery, and molecular pathway manipulation.


Subject(s)
Cell Lineage/genetics , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Myocytes, Cardiac/cytology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Transdifferentiation/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Small Molecule Libraries/pharmacology , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
7.
Biomaterials ; 85: 180-194, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26874281

ABSTRACT

Access to smooth muscle cells (SMC) would create opportunities for tissue engineering, drug testing, and disease modeling. Herein we report the direct conversion of human endothelial progenitor cells (EPC) to induced smooth muscle cells (iSMC) by induced expression of MYOCD. The EPC undergo a cytoskeletal rearrangement resembling that of mesenchymal cells within 3 days post initiation of MYOCD expression. By day 7, the reprogrammed cells show upregulation of smooth muscle markers ACTA2, MYH11, and TAGLN by qRT-PCR and ACTA2 and MYH11 expression by immunofluorescence. By two weeks, they resemble umbilical artery SMC in microarray gene expression analysis. The iSMC, in contrast to EPC control, show calcium transients in response to phenylephrine stimulation and a contractility an order of magnitude higher than that of EPC as determined by traction force microscopy. Tissue-engineered blood vessels constructed using iSMC show functionality with respect to flow- and drug-mediated vasodilation and vasoconstriction.


Subject(s)
Cell Transdifferentiation , Endothelial Progenitor Cells/cytology , Myocytes, Smooth Muscle/cytology , Actins/genetics , Actins/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Down-Regulation , Endoglin/genetics , Endoglin/metabolism , Gene Transfer Techniques , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tissue Engineering , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation
8.
Acta Biomater ; 32: 100-109, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26708709

ABSTRACT

Burn injuries in the United States account for over one million hospital admissions per year, with treatment estimated at four billion dollars. Of severe burn patients, 30-90% will develop hypertrophic scars (HSc). In this study, we evaluate the impact of an elastomeric, randomly-oriented biostable polyurethane (PU) scaffold on HSc-related outcomes. In vitro, fibroblast-seeded PU scaffolds contracted significantly less and demonstrated fewer αSMA(+) myofibroblasts compared to fibroblast-seeded collagen lattices. In a murine HSc model, collagen coated PU (ccPU) scaffolds significantly reduced HSc contraction as compared to untreated control wounds and wounds treated with the clinical standard of care. Our data suggest that electrospun ccPU scaffolds meet the requirements to reduce HSc contraction including reduction of in vitro HSc related outcomes, diminished scar stiffness, and reduced scar contraction. While clinical dogma suggests treating severe burn patients with rapidly biodegrading skin equivalents, our data suggest that a more long-term scaffold may possess merit in reducing HSc. STATEMENT OF SIGNIFICANCE: In severe burns treated with skin grafting, between 30% and 90% of patients develop hypertrophic scars (HSc). There are no therapies to prevent HSc, and treatments are marginally effective. This work is the first example we are aware of which studies the impact of a permanent electrospun elastomer on HSc contraction in a murine model that mimics the human condition. Collagen coated polyurethane scaffolds decrease αSMA+ myofibroblast formation in vitro, prevent stiffening of scar tissue, and mitigate HSc contraction. Unlike current standards of care, electrospun, polyurethane scaffolds do not lose architecture over time. We propose that the future bioengineering strategy of mitigating HSc contraction should consider a long-term elastomeric matrix which persists within the wound bed throughout the remodeling phase of repair.


Subject(s)
Cicatrix, Hypertrophic/pathology , Immunocompetence , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Wound Healing , Animals , Biomechanical Phenomena , Cattle , Dermis/pathology , Disease Models, Animal , Female , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Mice, Inbred C57BL , Polyurethanes/chemistry
9.
Sci Rep ; 4: 7403, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25492703

ABSTRACT

Transgene insertion plays an important role in gene therapy and in biological studies. Transposon-based systems that integrate transgenes by transposase-catalyzed "cut-and-paste" mechanism have emerged as an attractive system for transgenesis. Hyperactive piggyBac transposon is particularly promising due to its ability to integrate large transgenes with high efficiency. However, prolonged expression of transposase can become a potential source of genotoxic effects due to uncontrolled transposition of the integrated transgene from one chromosomal locus to another. In this study we propose a vector design to decrease post-transposition expression of transposase and to eliminate the cells that have residual transposase expression. We design a single plasmid construct that combines the transposase and the transpositioning transgene element to share a single polyA sequence for termination. Consequently, the separation of the transposase element from the polyA sequence after transposition leads to its deactivation. We also co-express Herpes Simplex Virus thymidine kinase (HSV-tk) with the transposase. Therefore, cells having residual transposase expression can be eliminated by the administration of ganciclovir. We demonstrate the utility of this combination transposon system by integrating and expressing a model therapeutic gene, human coagulation Factor IX, in HEK293T cells.


Subject(s)
DNA Transposable Elements , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Factor IX/biosynthesis , Factor IX/genetics , HEK293 Cells , Humans , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics , Transposases/biosynthesis , Transposases/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Stem Cell Reports ; 3(6): 940-7, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25448066

ABSTRACT

Gene activation by the CRISPR/Cas9 system has the potential to enable new approaches to science and medicine, but the technology must be enhanced to robustly control cell behavior. We show that the fusion of two transactivation domains to Cas9 dramatically enhances gene activation to a level that is necessary to reprogram cell phenotype. Targeted activation of the endogenous Myod1 gene locus with this system led to stable and sustained reprogramming of mouse embryonic fibroblasts into skeletal myocytes. The levels of myogenic marker expression obtained by the activation of endogenous Myod1 gene were comparable to that achieved by overexpression of lentivirally delivered MYOD1 transcription factor.


Subject(s)
CRISPR-Cas Systems , Cell Lineage/genetics , Cellular Reprogramming , Animals , Cell Line , Gene Expression , Gene Expression Regulation , Mice , MyoD Protein/genetics , MyoD Protein/metabolism , Organ Specificity/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
PLoS One ; 8(6): e65963, 2013.
Article in English | MEDLINE | ID: mdl-23785459

ABSTRACT

The mammalian heart has little capacity to regenerate, and following injury the myocardium is replaced by non-contractile scar tissue. Consequently, increased wall stress and workload on the remaining myocardium leads to chamber dilation, dysfunction, and heart failure. Cell-based therapy with an autologous, epigenetically reprogrammed, and cardiac-committed progenitor cell source could potentially reverse this process by replacing the damaged myocardium with functional tissue. However, it is unclear whether cardiac progenitor cell-derived cardiomyocytes are capable of attaining levels of structural and functional maturity comparable to that of terminally-fated cardiomyocytes. Here, we first describe the derivation of mouse induced pluripotent stem (iPS) cells, which once differentiated allow for the enrichment of Nkx2-5(+) cardiac progenitors, and the cardiomyocyte-specific expression of the red fluorescent protein. We show that the cardiac progenitors are multipotent and capable of differentiating into endothelial cells, smooth muscle cells and cardiomyocytes. Moreover, cardiac progenitor selection corresponds to cKit(+) cell enrichment, while cardiomyocyte cell-lineage commitment is concomitant with dual expression of either cKit/Flk1 or cKit/Sca-1. We proceed to show that the cardiac progenitor-derived cardiomyocytes are capable of forming electrically and mechanically coupled large-scale 2D cell cultures with mature electrophysiological properties. Finally, we examine the cell progenitors' ability to form electromechanically coherent macroscopic tissues, using a physiologically relevant 3D culture model and demonstrate that following long-term culture the cardiomyocytes align, and form robust electromechanical connections throughout the volume of the biosynthetic tissue construct. We conclude that the iPS cell-derived cardiac progenitors are a robust cell source for tissue engineering applications and a 3D culture platform for pharmacological screening and drug development studies.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Action Potentials , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Gene Expression , Gene Expression Profiling , Immunophenotyping , Induced Pluripotent Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Tissue Engineering , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic
12.
PLoS One ; 8(5): e64342, 2013.
Article in English | MEDLINE | ID: mdl-23717601

ABSTRACT

Viral vectors remain the most efficient and popular in deriving induced pluripotent stem cells (iPSCs). For translation, it is important to silence or remove the reprogramming factors after induction of pluripotency. In this study, we design an excisable loxP-flanked lentiviral construct that a) includes all the reprogramming elements in a single lentiviral vector expressed by a strong EF-1α promoter; b) enables easy determination of lentiviral titer; c) enables transgene removal and cell enrichment using LoxP-site-specific Cre-recombinase excision and Herpes Simplex Virus-thymidine kinase/ganciclovir (HSV-tk/gan) negative selection; and d) allows for transgene excision in a colony format. A reprogramming efficiency comparable to that reported in the literature without boosting molecules can be consistently obtained. To further demonstrate the utility of this Cre-loxP/HSV-tk/gan strategy, we incorporate a non-viral therapeutic transgene (human blood coagulation Factor IX) in the iPSCs, whose expression can be controlled by a temporal pulse of Cre recombinase. The robustness of this platform enables the implementation of an efficacious and cost-effective protocol for iPSC generation and their subsequent transgenesis for downstream studies.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Integrases/metabolism , Recombination, Genetic , Transgenes , Animals , Antiviral Agents/pharmacology , Base Sequence , Cells, Cultured , DNA Primers , Ganciclovir/pharmacology , Mice , Polymerase Chain Reaction , Simplexvirus/enzymology , Thymidine Kinase/genetics
13.
Adv Drug Deliv Rev ; 61(12): 1043-54, 2009 Oct 05.
Article in English | MEDLINE | ID: mdl-19651167

ABSTRACT

Electrospinning and electrospraying are facile electrohydrodynamic fabrication methods that can generate drug delivery systems (DDS) through a one-step process. The nanostructured fiber and particle morphologies produced by these techniques offer tunable release kinetics applicable to diverse biomedical applications. Coaxial electrospinning/electrospraying, a relatively new technique of fabricating core-shell fibers/particles have added to the versatility of these DDS by affording a near zero-order drug release kinetics, dampening of burst release, and applicability to a wider range of bioactive agents. Controllable electrospinning/spraying of fibers and particles and subsequent drug release from these chiefly polymeric vehicles depends on well-defined solution and process parameters. The additional drug delivery capability from electrospun fibers can further enhance the material's functionality in tissue engineering applications. This review discusses the state-of-the-art of using electrohydrodynamic technique to generate nanofiber/particles as drug delivery devices.


Subject(s)
Drug Compounding/methods , Drug Delivery Systems/methods , Nanofibers/chemistry , Nanoparticles/chemistry , Pharmaceutical Preparations/administration & dosage , Drug Compounding/instrumentation , Electrochemistry , Models, Theoretical , Tissue Engineering
14.
J Biomed Mater Res B Appl Biomater ; 88(1): 17-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18386845

ABSTRACT

Electrospraying is a novel technique for the generation of micro/nanospheres for biomedical applications. Apart from being a high yield technique; electrospraying has an added advantage of not making use of an external dispersion/emulsion phase which often involves ingredients that are undesirable for biomedical applications. In this study, we report the use of electrospraying for the synthesis of chitosan micro/nanospheres. The focus was to optimize the fabrication parameters involved in electrospraying for reproducible synthesis of chitosan based micro/nanospheres and to study their potential as delivery vehicles for bioactive agents. The influence of the following was studied (i) electrospraying voltage, (ii) needle gauge, (iii) concentration of chitosan solution, (iv) concentration of acetic acid solution, and (v) electrospraying distance. SEM analysis demonstrated that microspheres of less than 1 mum were obtained when chitosan concentration was 2% dissolved in 90% acetic acid. The working distance and needle gauge that yielded favorable results were 7 cm and 26 g, respectively. Average particle size of ampicillin loaded chitosan micro/nanospheres was 520 nm with zeta potential of +28.2 mV and encapsulation efficiency of 80.4%. The particles were characterized for drug release kinetics and results demonstrated an initial burst release followed by a sustained release over a period of 120 h. Further, antibacterial activity of drug loaded micro/nanospheres demonstrated that the encapsulated drug was in its active form postexposure to high voltage during electrospraying. This study indicates that electrospraying is a facile technique for the synthesis of chitosan micro/nanospheres for drug delivery applications.


Subject(s)
Biocompatible Materials/chemistry , Chitosan/chemistry , Drug Carriers , Drug Delivery Systems , Nanospheres/chemistry , Nanotechnology/methods , Acetic Acid/chemistry , Anti-Bacterial Agents/administration & dosage , Drug Compounding/methods , Drug Design , Electrochemistry/methods , Microscopy, Electron, Scanning/methods , Microspheres , Particle Size , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...