62 results on '"Pyle AD"'
Search Results
2. Publisher Correction: Regenerating human skeletal muscle forms an emerging niche in vivo to support PAX7 cells.
- Author
-
Hicks MR, Saleh KK, Clock B, Gibbs DE, Yang M, Younesi S, Gane L, Gutierrez-Garcia V, Xi H, and Pyle AD
- Published
- 2024
- Full Text
- View/download PDF
3. 'Enhancing' skeletal muscle and stem cells in three-dimensions: genome regulation of skeletal muscle in development and disease.
- Author
-
Romero MA and Pyle AD
- Subjects
- Cell Differentiation genetics, Stem Cells metabolism, Enhancer Elements, Genetic, Transcription Factors genetics, Muscle, Skeletal metabolism
- Abstract
The noncoding genome imparts important regulatory control over gene expression. In particular, gene enhancers represent a critical layer of control that integrates developmental and differentiation signals outside the cell into transcriptional outputs inside the cell. Recently, there has been an explosion in genomic techniques to probe enhancer control, function, and regulation. How enhancers are regulated and integrate signals in stem cell development and differentiation is largely an open question. In this review, we focus on the role gene enhancers play in muscle stem cell specification, differentiation, and progression. We pay specific attention toward the identification of muscle-specific enhancers, the binding of transcription factors to these enhancers, and how enhancers communicate to their target genes via three-dimensional looping., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships that may be considered as potential competing interests: A.D.P. is cofounder and scientific advisor to MyoGene Bio in San Diego, CA., (Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
4. Regenerating human skeletal muscle forms an emerging niche in vivo to support PAX7 cells.
- Author
-
Hicks MR, Saleh KK, Clock B, Gibbs DE, Yang M, Younesi S, Gane L, Gutierrez-Garcia V, Xi H, and Pyle AD
- Subjects
- Animals, Humans, Mice, Pluripotent Stem Cells, Muscle, Skeletal physiology, PAX7 Transcription Factor genetics, PAX7 Transcription Factor metabolism, Satellite Cells, Skeletal Muscle physiology, Regeneration
- Abstract
Skeletal muscle stem and progenitor cells including those derived from human pluripotent stem cells (hPSCs) offer an avenue towards personalized therapies and readily fuse to form human-mouse myofibres in vivo. However, skeletal muscle progenitor cells (SMPCs) inefficiently colonize chimeric stem cell niches and instead associate with human myofibres resembling foetal niches. We hypothesized competition with mouse satellite cells (SCs) prevented SMPC engraftment into the SC niche and thus generated an SC ablation mouse compatible with human engraftment. Single-nucleus RNA sequencing of SC-ablated mice identified the absence of a transient myofibre subtype during regeneration expressing Actc1. Similarly, ACTC1
+ human myofibres supporting PAX7+ SMPCs increased in SC-ablated mice, and after re-injury we found SMPCs could now repopulate into chimeric niches. To demonstrate ACTC1+ myofibres are essential to supporting PAX7 SMPCs, we generated caspase-inducible ACTC1 depletion human pluripotent stem cells, and upon SMPC engraftment we found a 90% reduction in ACTC1+ myofibres and a 100-fold decrease in PAX7 cell numbers compared with non-induced controls. We used spatial RNA sequencing to identify key factors driving emerging human niche formation between ACTC1+ myofibres and PAX7+ SMPCs in vivo. This revealed that transient regenerating human myofibres are essential for emerging niche formation in vivo to support PAX7 SMPCs., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2023
- Full Text
- View/download PDF
5. Nanoparticles systemically biodistribute to regenerating skeletal muscle in DMD.
- Author
-
Hicks MR, Liu X, Young CS, Saleh K, Ji Y, Jiang J, Emami MR, Mokhonova E, Spencer MJ, Meng H, and Pyle AD
- Subjects
- Animals, Mice, Tissue Distribution, Mice, Inbred mdx, Regeneration, Dystrophin, Muscle, Skeletal
- Abstract
Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
6. SIX1+PAX3+ identify a progenitor for myogenic lineage commitment from hPSCs.
- Author
-
Jaime OG, Arias J, Pavani S, Pyle AD, and Hicks MR
- Subjects
- Adult, Humans, PAX3 Transcription Factor genetics, PAX3 Transcription Factor metabolism, Cell Differentiation genetics, Muscle, Skeletal metabolism, Muscle Development genetics, PAX7 Transcription Factor genetics, PAX7 Transcription Factor metabolism, Homeodomain Proteins metabolism, Pluripotent Stem Cells metabolism
- Abstract
The earliest skeletal muscle progenitor cells (SMPCs) derived from human pluripotent stem cells (hPSCs) are often identified by factors expressed by a diverse number of progenitors. An early transcriptional checkpoint that defines myogenic commitment could improve hPSC differentiation to skeletal muscle. Analysis of several myogenic factors in human embryos and early hPSC differentiations found SIX1+PAX3+ co-expression was most indictive of myogenesis. Using dCas9-KRAB hPSCs, we demonstrate that early inhibition of SIX1 alone significantly decreased PAX3 expression, reduced PAX7+ SMPCs, and myotubes later in differentiation. Emergence of SIX1+PAX3+ precursors can be improved by manipulating seeding density, monitoring metabolic secretion and altering the concentration of CHIR99021. These modifications resulted in the co-emergence of hPSC-derived sclerotome, cardiac and neural crest that we hypothesized enhanced hPSC myogenic differentiation. Inhibition of non-myogenic lineages modulated PAX3 independent of SIX1. To better understand SIX1 expression, we compared directed differentiations to fetal progenitors and adult satellite cells by RNA-seq. Although SIX1 continued to be expressed across human development, SIX1 co-factor expression was dependent on developmental timing. We provide a resource to enable efficient derivation of skeletal muscle from hPSCs., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
7. Innate and adaptive AAV-mediated immune responses in a mouse model of Duchenne muscular dystrophy.
- Author
-
Emami MR, Espinoza A, Young CS, Ma F, Farahat PK, Felgner PL, Chamberlain JS, Xu X, Pyle AD, Pellegrini M, Villalta SA, and Spencer MJ
- Abstract
High systemic doses of adeno-associated viruses (AAVs) have been associated with immune-related serious adverse events (SAEs). Although AAV was well tolerated in preclinical models, SAEs were observed in clinical trials, indicating the need for improved preclinical models to understand AAV-induced immune responses. Here, we show that mice dual-dosed with AAV9 at 4-week intervals better recapitulate aspects of human immunity to AAV. In the model, anti-AAV9 immunoglobulin G (IgGs) increased in a linear fashion between the first and second AAV administrations. Complement activation was only observed in the presence of high levels of both AAV and anti-AAV IgG. Myeloid-derived pro-inflammatory cytokines were significantly induced in the same pattern as complement activation, suggesting that myeloid cell activation to AAV may rely on the presence of both AAV and anti-AAV IgG complexes. Single-cell RNA sequencing of peripheral blood mononuclear cells confirmed that activated monocytes were a primary source of pro-inflammatory cytokines and chemokines, which were significantly increased after a second AAV9 exposure. The same activated monocyte clusters expressed both Fcγ and complement receptors, suggesting that anti-AAV-mediated activation of myeloid cells through Fcγ receptors and/or complement receptors is one mechanism by which anti-AAV antigen complexes may prime antigen-presenting cells and amplify downstream immunity., Competing Interests: M.J.S., A.D.P., and C.S.Y. are co-founders of MyoGene Bio, a startup spun out of UCLA developing gene editing therapies for Duchenne muscular dystrophy., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
8. Muscle fusogens go viral for gene delivery to skeletal muscle.
- Author
-
Gibbs DE and Pyle AD
- Subjects
- Animals, Cell Fusion, Mammals, Genetic Therapy, Muscle, Skeletal, Viruses
- Abstract
Viruses and multinucleated cells rely on fusogens to facilitate the fusion of their membranes. In this issue of Cell, Millay and colleagues demonstrate that replacing viral fusogens with mammalian skeletal muscle fusogens leads to the specific transduction of skeletal muscle and the ability to deliver gene therapy constructs in a therapeutically relevant muscle disease., Competing Interests: Declaration of interests A.D.P. is a co-founder and scientific advisor to MyoGene Bio, San Diego, CA, USA., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
9. Duchenne muscular dystrophy disease severity impacts skeletal muscle progenitor cells systemic delivery.
- Author
-
Saleh KK, Switzler C, Hicks MR, Gane L, Gibbs DE, and Pyle AD
- Abstract
Duchenne muscular dystrophy (DMD) is caused by an out-of-frame mutation in the DMD gene that results in the absence of a functional dystrophin protein, leading to a devastating progressive lethal muscle-wasting disease. Muscle stem cell-based therapy is a promising avenue for improving muscle regeneration. However, despite the efforts to deliver the optimal cell population to multiple muscles most efforts have failed. Here we describe a detailed optimized method of for the delivery of human skeletal muscle progenitor cells (SMPCs) to multiple hindlimb muscles in healthy, dystrophic and severely dystrophic mouse models. We show that systemic delivery is inefficient and is affected by the microenvironment. We found that significantly less human SMPCs were detected in healthy gastrocnemius muscle cross-sections, compared to both dystrophic and severely dystrophic gastrocnemius muscle. Human SMPCs were found to be detected inside blood vessels distinctly in healthy, dystrophic and severely dystrophic muscles, with prominent clotting identified in severely dystrophic muscles after intra arterial (IA) systemic cell delivery. We propose that muscle microenvironment and the severity of muscular dystrophy to an extent impacts the systemic delivery of SMPCs and that overall systemic stem cell delivery is not currently efficient or safe to be used in cell based therapies for DMD. This work extends our understanding of the severe nature of DMD, which should be taken into account when considering stem cell-based systemic delivery platforms., Competing Interests: AP is co-founder and SAB member of MyoGene Bio. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Saleh, Switzler, Hicks, Gane, Gibbs and Pyle.)
- Published
- 2023
- Full Text
- View/download PDF
10. Myoscaffolds reveal laminin scarring is detrimental for stem cell function while sarcospan induces compensatory fibrosis.
- Author
-
Stearns-Reider KM, Hicks MR, Hammond KG, Reynolds JC, Maity A, Kurmangaliyev YZ, Chin J, Stieg AZ, Geisse NA, Hohlbauch S, Kaemmer S, Schmitt LR, Pham TT, Yamauchi K, Novitch BG, Wollman R, Hansen KC, Pyle AD, and Crosbie RH
- Abstract
We developed an on-slide decellularization approach to generate acellular extracellular matrix (ECM) myoscaffolds that can be repopulated with various cell types to interrogate cell-ECM interactions. Using this platform, we investigated whether fibrotic ECM scarring affected human skeletal muscle progenitor cell (SMPC) functions that are essential for myoregeneration. SMPCs exhibited robust adhesion, motility, and differentiation on healthy muscle-derived myoscaffolds. All SPMC interactions with fibrotic myoscaffolds from dystrophic muscle were severely blunted including reduced motility rate and migration. Furthermore, SMPCs were unable to remodel laminin dense fibrotic scars within diseased myoscaffolds. Proteomics and structural analysis revealed that excessive collagen deposition alone is not pathological, and can be compensatory, as revealed by overexpression of sarcospan and its associated ECM receptors in dystrophic muscle. Our in vivo data also supported that ECM remodeling is important for SMPC engraftment and that fibrotic scars may represent one barrier to efficient cell therapy., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. The emergence of the stem cell niche.
- Author
-
Hicks MR and Pyle AD
- Subjects
- Humans, Cell Differentiation, Stem Cell Niche physiology, Muscle, Skeletal, Satellite Cells, Skeletal Muscle, Pluripotent Stem Cells
- Abstract
Stem cell niches are composed of dynamic microenvironments that support stem cells over a lifetime. The emerging niche is distinct from the adult because its main role is to support the progenitors that build organ systems in development. Emerging niches mature through distinct stages to form the adult niche and enable proper stem cell support. As a model of emerging niches, this review highlights how differences in the skeletal muscle microenvironment influence emerging versus satellite cell (SC) niche formation in skeletal muscle, which is among the most regenerative tissue systems. We contrast how stem cell niches regulate intrinsic properties between progenitor and stem cells throughout development to adulthood. We describe new applications for generating emerging niches from human pluripotent stem cells (hPSCs) using developmental principles and highlight potential applications for regeneration and therapeutics., Competing Interests: Declaration of interests Dr. Pyle is co-founder and scientific advisory board member of MyoGene Bio., (Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
12. Single cell sequencing maps skeletal muscle cellular diversity as disease severity increases in dystrophic mouse models.
- Author
-
Saleh KK, Xi H, Switzler C, Skuratovsky E, Romero MA, Chien P, Gibbs D, Gane L, Hicks MR, Spencer MJ, and Pyle AD
- Abstract
Duchenne muscular dystrophy (DMD) is caused by out-of-frame mutations in the DMD gene resulting in the absence of a functional dystrophin protein, leading to a devastating and progressive lethal muscle-wasting disease. Little is known about cellular heterogeneity as disease severity increases. Advances in single-cell RNA sequencing (scRNA-seq) enabled us to explore skeletal muscle-resident cell populations in healthy, dystrophic, and severely dystrophic mouse models. We found increased frequencies of activated fibroblasts, fibro-adipogenic progenitor cells, and pro-inflammatory macrophages in dystrophic gastrocnemius muscles and an upregulation of extracellular matrix genes on endothelial cells in dystrophic and severely dystrophic muscles. We observed a pronounced risk of clotting, especially in the severely dystrophic mice with increased expression of plasminogen activator inhibitor-1 in endothelial cells, indicating endothelial cell impairment as disease severity increases. This work extends our understanding of the severe nature of DMD which should be considered when developing single or combinatorial approaches for DMD., Competing Interests: Dr. Pyle and Dr. Spencer are co-founders and scientific advisory board members to MyoGene Bio. Dr. Pyle has a financial interest in MyoGene Bio and The UC Regents have licensed IP that I invented to MyoGene Bio.
- Published
- 2022
- Full Text
- View/download PDF
13. Single-cell analysis and functional characterization uncover the stem cell hierarchies and developmental origins of rhabdomyosarcoma.
- Author
-
Wei Y, Qin Q, Yan C, Hayes MN, Garcia SP, Xi H, Do D, Jin AH, Eng TC, McCarthy KM, Adhikari A, Onozato ML, Spentzos D, Neilsen GP, Iafrate AJ, Wexler LH, Pyle AD, Suvà ML, Dela Cruz F, Pinello L, and Langenau DM
- Subjects
- Child, Humans, Muscle, Skeletal pathology, Single-Cell Analysis, Stem Cells pathology, Rhabdomyosarcoma genetics, Rhabdomyosarcoma, Embryonal
- Abstract
Rhabdomyosarcoma (RMS) is a common childhood cancer that shares features with developing skeletal muscle. Yet, the conservation of cellular hierarchy with human muscle development and the identification of molecularly defined tumor-propagating cells has not been reported. Using single-cell RNA-sequencing, DNA-barcode cell fate mapping and functional stem cell assays, we uncovered shared tumor cell hierarchies in RMS and human muscle development. We also identified common developmental stages at which tumor cells become arrested. Fusion-negative RMS cells resemble early myogenic cells found in embryonic and fetal development, while fusion-positive RMS cells express a highly specific gene program found in muscle cells transiting from embryonic to fetal development at 7-7.75 weeks of age. Fusion-positive RMS cells also have neural pathway-enriched states, suggesting less-rigid adherence to muscle-lineage hierarchies. Finally, we identified a molecularly defined tumor-propagating subpopulation in fusion-negative RMS that shares remarkable similarity to bi-potent, muscle mesenchyme progenitors that can make both muscle and osteogenic cells., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
14. Recapitulating human myogenesis ex vivo using human pluripotent stem cells.
- Author
-
Chien P, Xi H, and Pyle AD
- Subjects
- Cell Differentiation physiology, Humans, Models, Biological, Muscle Development genetics, Muscle, Skeletal cytology, Muscle, Skeletal physiology, Myoblasts, Skeletal cytology, Myoblasts, Skeletal physiology, PAX7 Transcription Factor genetics, PAX7 Transcription Factor metabolism, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle physiology, Muscle Development physiology, Pluripotent Stem Cells cytology, Pluripotent Stem Cells physiology
- Abstract
Human pluripotent stem cells (hPSCs) provide a human model for developmental myogenesis, disease modeling and development of therapeutics. Differentiation of hPSCs into muscle stem cells has the potential to provide a cell-based therapy for many skeletal muscle wasting diseases. This review describes the current state of hPSCs towards recapitulating human myogenesis ex vivo, considerations of stem cell and progenitor cell state as well as function for future use of hPSC-derived muscle cells in regenerative medicine., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
15. CRISPR-Mediated Genomic Addition to CPS1 Deficient iPSCs is Insufficient to Restore Nitrogen Homeostasis.
- Author
-
Nitzahn M, Truong B, Khoja S, Vega-Crespo A, Le C, Eliav A, Makris G, Pyle AD, Häberle J, and Lipshutz GS
- Subjects
- Carbamoyl-Phosphate Synthase (Ammonia) genetics, Carbamoyl-Phosphate Synthase (Ammonia) metabolism, Clustered Regularly Interspaced Short Palindromic Repeats, Genomics, Homeostasis, Humans, Nitrogen, Induced Pluripotent Stem Cells
- Abstract
CPS1 deficiency is an inborn error of metabolism caused by loss-of-function mutations in the CPS1 gene, catalyzing the initial reaction of the urea cycle. Deficiency typically leads to toxic levels of plasma ammonia, cerebral edema, coma, and death, with the only curative treatment being liver transplantation; due to limited donor availability and the invasiveness and complications of the procedure, however, alternative therapies are needed. Induced pluripotent stem cells offer an alternative cell source to partial or whole liver grafts that theoretically would not require immune suppression regimens and additionally are amenable to genetic modifications. Here, we genetically modified CPS1 deficient patient-derived stem cells to constitutively express human codon optimized CPS1 from the AAVS1 safe harbor site. While edited stem cells efficiently differentiated to hepatocyte-like cells, they failed to metabolize ammonia more efficiently than their unedited counterparts. This unexpected result appears to have arisen in part due to transgene promoter methylation, and thus transcriptional silencing, in undifferentiated cells, impacting their capacity to restore the complete urea cycle function upon differentiation. As pluripotent stem cell strategies are being expanded widely for potential cell therapies, these results highlight the need for strict quality control and functional analysis to ensure the integrity of cell products., (Copyright ©2021, Yale Journal of Biology and Medicine.)
- Published
- 2021
16. Long-term repair of porcine articular cartilage using cryopreservable, clinically compatible human embryonic stem cell-derived chondrocytes.
- Author
-
Petrigliano FA, Liu NQ, Lee S, Tassey J, Sarkar A, Lin Y, Li L, Yu Y, Geng D, Zhang J, Shkhyan R, Bogdanov J, Van Handel B, Ferguson GB, Lee Y, Hinderer S, Tseng KC, Kavanaugh A, Crump JG, Pyle AD, Schenke-Layland K, Billi F, Wang L, Lieberman J, Hurtig M, and Evseenko D
- Abstract
Osteoarthritis (OA) impacts hundreds of millions of people worldwide, with those affected incurring significant physical and financial burdens. Injuries such as focal defects to the articular surface are a major contributing risk factor for the development of OA. Current cartilage repair strategies are moderately effective at reducing pain but often replace damaged tissue with biomechanically inferior fibrocartilage. Here we describe the development, transcriptomic ontogenetic characterization and quality assessment at the single cell level, as well as the scaled manufacturing of an allogeneic human pluripotent stem cell-derived articular chondrocyte formulation that exhibits long-term functional repair of porcine articular cartilage. These results define a new potential clinical paradigm for articular cartilage repair and mitigation of the associated risk of OA., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
17. Symmetry breaking of tissue mechanics in wound induced hair follicle regeneration of laboratory and spiny mice.
- Author
-
Harn HI, Wang SP, Lai YC, Van Handel B, Liang YC, Tsai S, Schiessl IM, Sarkar A, Xi H, Hughes M, Kaemmer S, Tang MJ, Peti-Peterdi J, Pyle AD, Woolley TE, Evseenko D, Jiang TX, and Chuong CM
- Subjects
- Animals, Epidermis physiology, Gene Expression Profiling, Hair Follicle anatomy & histology, Hair Follicle physiology, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microarray Analysis, Microscopy, Atomic Force, Models, Psychological, Morphogenesis genetics, Murinae, RNA-Seq, Regeneration genetics, Regenerative Medicine, Signal Transduction genetics, Signal Transduction physiology, Spatio-Temporal Analysis, Twist-Related Protein 1 genetics, Wound Healing genetics, Epidermis anatomy & histology, Epidermis metabolism, Hair Follicle metabolism, Morphogenesis physiology, Regeneration physiology, Twist-Related Protein 1 metabolism, Wound Healing physiology
- Abstract
Tissue regeneration is a process that recapitulates and restores organ structure and function. Although previous studies have demonstrated wound-induced hair neogenesis (WIHN) in laboratory mice (Mus), the regeneration is limited to the center of the wound unlike those observed in African spiny (Acomys) mice. Tissue mechanics have been implicated as an integral part of tissue morphogenesis. Here, we use the WIHN model to investigate the mechanical and molecular responses of laboratory and African spiny mice, and report these models demonstrate opposing trends in spatiotemporal morphogenetic field formation with association to wound stiffness landscapes. Transcriptome analysis and K14-Cre-Twist1 transgenic mice show the Twist1 pathway acts as a mediator for both epidermal-dermal interactions and a competence factor for periodic patterning, differing from those used in development. We propose a Turing model based on tissue stiffness that supports a two-scale tissue mechanics process: (1) establishing a morphogenetic field within the wound bed (mm scale) and (2) symmetry breaking of the epidermis and forming periodically arranged hair primordia within the morphogenetic field (μm scale). Thus, we delineate distinct chemo-mechanical events in building a Turing morphogenesis-competent field during WIHN of laboratory and African spiny mice and identify its evo-devo advantages with perspectives for regenerative medicine.
- Published
- 2021
- Full Text
- View/download PDF
18. Generation of PAX7 Reporter Cells to Investigate Skeletal Myogenesis from Human Pluripotent Stem Cells.
- Author
-
Xi H, Young CS, and Pyle AD
- Subjects
- 3' Untranslated Regions genetics, Amino Acid Sequence, Animals, Base Sequence, Binding Sites, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems genetics, Cell Count, Cell Differentiation, Conserved Sequence, Drug Resistance, Microbial, Genotype, Humans, Mammals, Mesoderm embryology, MicroRNAs genetics, MicroRNAs metabolism, PAX7 Transcription Factor chemistry, Plasmids genetics, Protein Isoforms chemistry, Protein Isoforms metabolism, RNA, Guide, CRISPR-Cas Systems genetics, Reproducibility of Results, Somites embryology, Genes, Reporter, Muscle Development, PAX7 Transcription Factor metabolism, Pluripotent Stem Cells metabolism
- Abstract
This protocol describes the use of CRISPR/Cas9-mediated homology-directed recombination to construct a PAX7-GFP reporter in human pluripotent stem cells (hPSCs). PAX7 is a key transcription factor and regulator of skeletal muscle stem/progenitor cells. We obtained heterozygous knockin reporter cells and validated their PAX7 expression using both artificial activation by the CRISPR/dCas9-VPR system and physiological activation during hPSC myogenic differentiation. These cells can serve as tools for better understanding of in vitro hPSC myogenesis and enriching myogenic cells for downstream analysis. For complete details on the use and execution of this protocol, please refer to Xi et al. (2017) and Xi et al. (2020)., Competing Interests: C.S.Y. and A.D.P. are co-founders of and have financial interests in MyoGene Bio. The Regents of the University of California have licensed intellectual property invented by C.S.Y. and A.D.P. to MyoGene Bio. A.D.P. serves on the scientific advisory board of MyoGene Bio. C.S.Y. is currently CEO of MyoGene Bio. H.X. declares no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
19. A Small-Molecule Approach to Restore a Slow-Oxidative Phenotype and Defective CaMKIIβ Signaling in Limb Girdle Muscular Dystrophy.
- Author
-
Liu J, Campagna J, John V, Damoiseaux R, Mokhonova E, Becerra D, Meng H, McNally EM, Pyle AD, Kramerova I, and Spencer MJ
- Subjects
- Acyltransferases genetics, Acyltransferases metabolism, Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Calpain deficiency, Cardiac Myosins metabolism, Cell Line, Creatine Kinase, Mitochondrial Form genetics, Creatine Kinase, Mitochondrial Form metabolism, Female, Gene Expression Regulation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Proteins deficiency, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Dystrophies, Limb-Girdle genetics, Muscular Dystrophies, Limb-Girdle metabolism, Muscular Dystrophies, Limb-Girdle pathology, Myoblasts drug effects, Myoblasts metabolism, Myoblasts pathology, Myosin Light Chains metabolism, Oxidative Stress, Phenotype, Physical Conditioning, Animal, Protein Isoforms genetics, Protein Isoforms metabolism, Signal Transduction, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Calpain genetics, Cardiac Myosins genetics, Muscle Proteins genetics, Muscular Dystrophies, Limb-Girdle drug therapy, Myosin Light Chains genetics, Pyrimidines pharmacology, Small Molecule Libraries pharmacology
- Abstract
Mutations in CAPN3 cause limb girdle muscular dystrophy R1 (LGMDR1, formerly LGMD2A) and lead to progressive and debilitating muscle wasting. Calpain 3 deficiency is associated with impaired CaMKIIβ signaling and blunted transcriptional programs that encode the slow-oxidative muscle phenotype. We conducted a high-throughput screen on a target of CaMKII ( Myl2 ) to identify compounds to override this signaling defect; 4 were tested in vivo in the Capn3 knockout (C3KO) model of LGMDR1. The leading compound, AMBMP, showed good exposure and was able to reverse the LGMDR1 phenotype in vivo , including improved oxidative properties, increased slow fiber size, and enhanced exercise performance. AMBMP also activated CaMKIIβ signaling, but it did not alter other pathways known to be associated with muscle growth. Thus, AMBMP treatment activates CaMKII and metabolically reprograms skeletal muscle toward a slow muscle phenotype. These proof-of-concept studies lend support for an approach to the development of therapeutics for LGMDR1., Competing Interests: M.J.S. and A.D.P. are co-founders of MyoGene Bio and are members of its scientific advisory board. M.J.S., I.K., J.C., and V.J. are inventors on a patent pending pertaining to new chemical entities of AMBMP., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
20. Genome Editing-Mediated Utrophin Upregulation in Duchenne Muscular Dystrophy Stem Cells.
- Author
-
Sengupta K, Mishra MK, Loro E, Spencer MJ, Pyle AD, and Khurana TS
- Abstract
Utrophin upregulation is considered a promising therapeutic strategy for Duchenne muscular dystrophy (DMD). A number of microRNAs (miRNAs) post-transcriptionally regulate utrophin expression by binding their cognate sites in the 3' UTR. Previously we have shown that miRNA: UTRN repression can be alleviated using miRNA let-7c site blocking oligonucleotides (SBOs) to achieve utrophin upregulation and functional improvement in mdx mice. Here, we used CRISPR/Cas9-mediated genome editing to delete five miRNA binding sites (miR-150, miR-296-5p, miR-133b, let-7c, miR-196b) clustered in a 500 bp inhibitory miRNA target region (IMTR) within the UTRN 3' UTR, for achieving higher expression of endogenous utrophin. Deleting the UTRN IMTR in DMD patient-derived human induced pluripotent stem cells (DMD-hiPSCs) resulted in ca. 2-fold higher levels of utrophin protein. Differentiation of the UTRN edited DMD-hiPSCs ( UTRN ΔIMTR) by MyoD overexpression resulted in increased sarcolemmal α-sarcoglycan staining consistent with improved dystrophin glycoprotein complex (DGC) restoration. These results demonstrate that CRISPR/Cas9-based UTRN genome editing offers a novel utrophin upregulation therapeutic strategy applicable to all DMD patients, irrespective of the dystrophin mutation status., (© 2020.)
- Published
- 2020
- Full Text
- View/download PDF
21. A Human Skeletal Muscle Atlas Identifies the Trajectories of Stem and Progenitor Cells across Development and from Human Pluripotent Stem Cells.
- Author
-
Xi H, Langerman J, Sabri S, Chien P, Young CS, Younesi S, Hicks M, Gonzalez K, Fujiwara W, Marzi J, Liebscher S, Spencer M, Van Handel B, Evseenko D, Schenke-Layland K, Plath K, and Pyle AD
- Published
- 2020
- Full Text
- View/download PDF
22. Non-fibro-adipogenic pericytes from human embryonic stem cells attenuate degeneration of the chronically injured mouse muscle.
- Author
-
Mosich GM, Husman R, Shah P, Sharma A, Rezzadeh K, Aderibigbe T, Hu VJ, McClintick DJ, Wu G, Gatto JD, Xi H, Pyle AD, Péault B, Petrigliano FA, and Dar A
- Subjects
- Animals, Cell Differentiation, Cell Line, Chronic Disease therapy, Disease Models, Animal, Female, Fibrosis, Humans, Injections, Intralesional, Mice, Muscle Development physiology, Muscular Disorders, Atrophic etiology, Muscular Disorders, Atrophic pathology, Muscular Disorders, Atrophic physiopathology, Pericytes physiology, Rotator Cuff physiopathology, Transplantation, Heterologous methods, Human Embryonic Stem Cells physiology, Muscular Disorders, Atrophic therapy, Pericytes transplantation, Rotator Cuff pathology, Rotator Cuff Injuries complications
- Abstract
Massive tears of the rotator cuff (RC) are associated with chronic muscle degeneration due to fibrosis, fatty infiltration, and muscle atrophy. The microenvironment of diseased muscle often impairs efficient engraftment and regenerative activity of transplanted myogenic precursors. Accumulating myofibroblasts and fat cells disrupt the muscle stem cell niche and myogenic cell signaling and deposit excess disorganized connective tissue. Therefore, restoration of the damaged stromal niche with non-fibro-adipogenic cells is a prerequisite to successful repair of an injured RC. We generated from human embryonic stem cells (hES) a potentially novel subset of PDGFR-β+CD146+CD34-CD56- pericytes that lack expression of the fibro-adipogenic cell marker PDGFR-α. Accordingly, the PDGFR-β+PDGFR-α- phenotype typified non-fibro-adipogenic, non-myogenic, pericyte-like derivatives that maintained non-fibro-adipogenic properties when transplanted into chronically injured murine RCs. Although administered hES pericytes inhibited developing fibrosis at early and late stages of progressive muscle degeneration, transplanted PDGFR-β+PDGFR-α+ human muscle-derived fibro-adipogenic progenitors contributed to adipogenesis and greater fibrosis. Additionally, transplanted hES pericytes substantially attenuated muscle atrophy at all tested injection time points after injury. Coinciding with this observation, conditioned medium from cultured hES pericytes rescued atrophic myotubes in vitro. These findings imply that non-fibro-adipogenic hES pericytes recapitulate the myogenic stromal niche and may be used to improve cell-based treatments for chronic muscle disorders.
- Published
- 2019
- Full Text
- View/download PDF
23. A customizable microfluidic platform for medium-throughput modeling of neuromuscular circuits.
- Author
-
Bellmann J, Goswami RY, Girardo S, Rein N, Hosseinzadeh Z, Hicks MR, Busskamp V, Pyle AD, Werner C, and Sterneckert J
- Subjects
- Cell Adhesion drug effects, Cell Differentiation drug effects, Cells, Cultured, Dimethylpolysiloxanes chemistry, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells drug effects, Laminin pharmacology, Maleates chemistry, Motor Neurons cytology, Motor Neurons drug effects, Muscle Fibers, Skeletal cytology, Muscle Fibers, Skeletal drug effects, Peptides pharmacology, Plasma Gases chemistry, Polyethylenes chemistry, Microfluidics methods, Neuromuscular Junction physiology
- Abstract
Neuromuscular circuits (NMCs) are vital for voluntary movement, and effective models of NMCs are needed to understand the pathogenesis of, as well as to identify effective treatments for, multiple diseases, including Duchenne's muscular dystrophy and amyotrophic lateral sclerosis. Microfluidics are ideal for recapitulating the central and peripheral compartments of NMCs, but myotubes often detach before functional NMCs are formed. In addition, microfluidic systems are often limited to a single experimental unit, which significantly limits their application in disease modeling and drug discovery. Here, we developed a microfluidic platform (MFP) containing over 100 experimental units, making it suitable for medium-throughput applications. To overcome detachment, we incorporated a reactive polymer surface allowing customization of the environment to culture different cell types. Using this approach, we identified conditions that enable long-term co-culture of human motor neurons and myotubes differentiated from human induced pluripotent stem cells inside our MFP. Optogenetics demonstrated the formation of functional NMCs. Furthermore, we developed a novel application of the rabies tracing assay to efficiently identify NMCs in our MFP. Therefore, our MFP enables large-scale generation and quantification of functional NMCs for disease modeling and pharmacological drug targeting., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
24. CRISPR for Neuromuscular Disorders: Gene Editing and Beyond.
- Author
-
Young CS, Pyle AD, and Spencer MJ
- Subjects
- Animals, Gene Editing methods, Humans, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Neuromuscular Diseases genetics
- Abstract
This is a review describing advances in CRISPR/Cas-mediated therapies for neuromuscular disorders (NMDs). We explore both CRISPR-mediated editing and dead Cas approaches as potential therapeutic strategies for multiple NMDs. Last, therapeutic considerations, including delivery and off-target effects, are also discussed.
- Published
- 2019
- Full Text
- View/download PDF
25. Correction to: Differentiation of RPE cells from integration-free iPS cells and their cell biological characterization.
- Author
-
Hazim RA, Karumbayaram S, Jiang M, Dimashkie A, Lopes VS, Li D, Burgess BL, Vijayaraj P, Alva-Ornelas JA, Zack JA, Kohn DB, Gomperts BN, Pyle AD, Lowry WE, and Williams DS
- Abstract
The original article [1] contains an error in the legend of Fig 5 whereby the descriptions for panels 5d and 5e are incorrect; as such, the corrected legend can be viewed below with its respective figure images.
- Published
- 2019
- Full Text
- View/download PDF
26. Mapping molecular landmarks of human skeletal ontogeny and pluripotent stem cell-derived articular chondrocytes.
- Author
-
Ferguson GB, Van Handel B, Bay M, Fiziev P, Org T, Lee S, Shkhyan R, Banks NW, Scheinberg M, Wu L, Saitta B, Elphingstone J, Larson AN, Riester SM, Pyle AD, Bernthal NM, Mikkola HK, Ernst J, van Wijnen AJ, Bonaguidi M, and Evseenko D
- Subjects
- Animals, Biomarkers metabolism, Epigenesis, Genetic, Fetal Development, Gene Expression Profiling, Histone Code, Humans, Mice, Sequence Analysis, RNA, Swine, Transcription, Genetic, Transcriptome, Chondrocytes metabolism, Chondrogenesis, Myoblasts metabolism, Osteoblasts metabolism, Tenocytes metabolism
- Abstract
Tissue-specific gene expression defines cellular identity and function, but knowledge of early human development is limited, hampering application of cell-based therapies. Here we profiled 5 distinct cell types at a single fetal stage, as well as chondrocytes at 4 stages in vivo and 2 stages during in vitro differentiation. Network analysis delineated five tissue-specific gene modules; these modules and chromatin state analysis defined broad similarities in gene expression during cartilage specification and maturation in vitro and in vivo, including early expression and progressive silencing of muscle- and bone-specific genes. Finally, ontogenetic analysis of freshly isolated and pluripotent stem cell-derived articular chondrocytes identified that integrin alpha 4 defines 2 subsets of functionally and molecularly distinct chondrocytes characterized by their gene expression, osteochondral potential in vitro and proliferative signature in vivo. These analyses provide new insight into human musculoskeletal development and provide an essential comparative resource for disease modeling and regenerative medicine.
- Published
- 2018
- Full Text
- View/download PDF
27. Suicide Prevention for School Communities: An Educational Initiative for Student Safety.
- Author
-
Roberts DC, Taylor ME, and Pyle AD
- Subjects
- Adolescent, Child, Humans, Texas, Health Education, Nurse's Role, School Nursing, Suicide Prevention
- Abstract
A knowledge gap exists in school communities regarding suicide prevention and means reduction education. The article highlights two core interrelated topics: school nurse engagement in dialogue with students' families and the implementation of an innovative, community-based suicide prevention educational program at a suburban public school district. The authors provide an overview of the public health problem of suicide for students, current student challenges, role of the school nurse in suicide prevention, and a key gap in current school nursing practice. At the request of the school counselors and principal, an innovative suicide prevention educational program was initiated as a community-based project at a large suburban public school district in Texas. The two overarching goals for this community-based collaboration are the following: school nurses will engage in frank, productive conversations with students' parents and families about suicidality concerns and increase the school community's knowledge about suicide prevention. This school community knowledge includes effective risk mitigation and means reduction strategies to better manage suicidality in students. Ultimately, this ongoing family and school community collaboration aims to prevent student deaths by suicide.
- Published
- 2018
- Full Text
- View/download PDF
28. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs.
- Author
-
Hicks MR, Hiserodt J, Paras K, Fujiwara W, Eskin A, Jan M, Xi H, Young CS, Evseenko D, Nelson SF, Spencer MJ, Handel BV, and Pyle AD
- Subjects
- Adult, Aged, CRISPR-Cas Systems, Cell Differentiation, Dystrophin genetics, Dystrophin metabolism, Female, Gene Editing, Gene Expression Regulation, Developmental, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Male, Middle Aged, Muscle Fibers, Skeletal cytology, Muscular Dystrophy, Duchenne metabolism, Muscular Dystrophy, Duchenne pathology, Muscular Dystrophy, Duchenne therapy, Myoblasts cytology, Myosins genetics, Myosins metabolism, Nerve Tissue Proteins metabolism, PAX7 Transcription Factor genetics, PAX7 Transcription Factor metabolism, Receptor, ErbB-3 metabolism, Receptors, Nerve Growth Factor metabolism, Signal Transduction, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Muscle Development genetics, Muscle Fibers, Skeletal metabolism, Muscular Dystrophy, Duchenne genetics, Myoblasts metabolism, Nerve Tissue Proteins genetics, Receptor, ErbB-3 genetics, Receptors, Nerve Growth Factor genetics
- Abstract
Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-β signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells.
- Published
- 2018
- Full Text
- View/download PDF
29. Differentiation of RPE cells from integration-free iPS cells and their cell biological characterization.
- Author
-
Hazim RA, Karumbayaram S, Jiang M, Dimashkie A, Lopes VS, Li D, Burgess BL, Vijayaraj P, Alva-Ornelas JA, Zack JA, Kohn DB, Gomperts BN, Pyle AD, Lowry WE, and Williams DS
- Subjects
- Animals, Cell Differentiation drug effects, Cell Polarity drug effects, Disease Models, Animal, Encephalitis Virus, Venezuelan Equine metabolism, Epithelial Cells cytology, Epithelial Cells physiology, Epithelial Cells transplantation, Fibroblasts cytology, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells drug effects, Injections, Intraocular, Intercellular Signaling Peptides and Proteins pharmacology, Mice, Mice, Inbred BALB C, Mice, Knockout, Primary Cell Culture, Retinal Degeneration pathology, Retinal Degeneration physiopathology, Retinal Pigment Epithelium cytology, Retinal Pigment Epithelium drug effects, Retinal Pigment Epithelium physiology, Skin cytology, Cellular Reprogramming genetics, Encephalitis Virus, Venezuelan Equine genetics, Epithelial Cells drug effects, Fibroblasts physiology, Induced Pluripotent Stem Cells physiology, Retinal Degeneration therapy
- Abstract
Background: Dysfunction of the retinal pigment epithelium (RPE) is implicated in numerous forms of retinal degeneration. The readily accessible environment of the eye makes it particularly suitable for the transplantation of RPE cells, which can now be derived from autologous induced pluripotent stem cells (iPSCs), to treat retinal degeneration. For RPE transplantation to become feasible in the clinic, patient-specific somatic cells should be reprogrammed to iPSCs without the introduction of reprogramming genes into the genome of the host cell, and then subsequently differentiated into RPE cells that are well characterized for safety and functionality prior to transplantation., Methods: We have reprogrammed human dermal fibroblasts to iPSCs using nonintegrating RNA, and differentiated the iPSCs toward an RPE fate (iPSC-RPE), under Good Manufacturing Practice (GMP)-compatible conditions., Results: Using highly sensitive assays for cell polarity, structure, organelle trafficking, and function, we found that iPSC-RPE cells in culture exhibited key characteristics of native RPE. Importantly, we demonstrate for the first time with any stem cell-derived RPE cell that live cells are able to support dynamic organelle transport. This highly sensitive test is critical for RPE cells intended for transplantation, since defects in intracellular motility have been shown to promote RPE pathogenesis akin to that found in macular degeneration. To test their capabilities for in-vivo transplantation, we injected the iPSC-RPE cells into the subretinal space of a mouse model of retinal degeneration, and demonstrated that the transplanted cells are capable of rescuing lost RPE function., Conclusions: This report documents the successful generation, under GMP-compatible conditions, of human iPSC-RPE cells that possess specific characteristics of healthy RPE. The report adds to a growing literature on the utility of human iPSC-RPE cells for cell culture investigations on pathogenicity and for therapeutic transplantation, by corroborating findings of others, and providing important new information on essential RPE cell biological properties.
- Published
- 2017
- Full Text
- View/download PDF
30. In Vivo Human Somitogenesis Guides Somite Development from hPSCs.
- Author
-
Xi H, Fujiwara W, Gonzalez K, Jan M, Liebscher S, Van Handel B, Schenke-Layland K, and Pyle AD
- Subjects
- Body Patterning physiology, Cell Differentiation physiology, Cells, Cultured, Gene Expression Regulation, Developmental physiology, Humans, Mesoderm metabolism, Mesoderm physiology, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Pluripotent Stem Cells metabolism, Signal Transduction physiology, Somites metabolism, Transforming Growth Factor beta metabolism, beta Catenin metabolism, Embryonic Development physiology, Morphogenesis physiology, Pluripotent Stem Cells physiology, Somites physiology
- Abstract
Somites form during embryonic development and give rise to unique cell and tissue types, such as skeletal muscles and bones and cartilage of the vertebrae. Using somitogenesis-stage human embryos, we performed transcriptomic profiling of human presomitic mesoderm as well as nascent and developed somites. In addition to conserved pathways such as WNT-β-catenin, we also identified BMP and transforming growth factor β (TGF-β) signaling as major regulators unique to human somitogenesis. This information enabled us to develop an efficient protocol to derive somite cells in vitro from human pluripotent stem cells (hPSCs). Importantly, the in-vitro-differentiating cells progressively expressed markers of the distinct developmental stages that are known to occur during in vivo somitogenesis. Furthermore, when subjected to lineage-specific differentiation conditions, the hPSC-derived somite cells were multipotent in generating somite derivatives, including skeletal myocytes, osteocytes, and chondrocytes. This work improves our understanding of human somitogenesis and may enhance our ability to treat diseases affecting somite derivatives., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. Creation of a Novel Humanized Dystrophic Mouse Model of Duchenne Muscular Dystrophy and Application of a CRISPR/Cas9 Gene Editing Therapy.
- Author
-
Young CS, Mokhonova E, Quinonez M, Pyle AD, and Spencer MJ
- Subjects
- Animals, CRISPR-Cas Systems, Dystrophin metabolism, Exons, Gene Editing methods, HEK293 Cells, Humans, Mice, Transgenic, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne metabolism, Muscular Dystrophy, Duchenne pathology, Disease Models, Animal, Dystrophin genetics, Genetic Therapy methods, Muscular Dystrophy, Duchenne therapy
- Abstract
Duchenne muscular dystrophy is caused by mutations in DMD which disrupt the reading frame. Therapeutic strategies that restore DMD's reading frame, such as exon skipping and CRISPR/Cas9, need to be tested in the context of the human DMD sequence in vivo. We have developed a novel dystrophic mouse model by using CRISPR/Cas9 to delete exon 45 in the human DMD gene in hDMD mice, which places DMD out-of-frame. We have utilized this model to demonstrate that our clinically-relevant CRISPR/Cas9 platform, which targets deletion of human DMD exons 45-55, can be directly applied in vivo to restore dystrophin.
- Published
- 2017
- Full Text
- View/download PDF
32. Restoring Ureagenesis in Hepatocytes by CRISPR/Cas9-mediated Genomic Addition to Arginase-deficient Induced Pluripotent Stem Cells.
- Author
-
Lee PC, Truong B, Vega-Crespo A, Gilmore WB, Hermann K, Angarita SA, Tang JK, Chang KM, Wininger AE, Lam AK, Schoenberg BE, Cederbaum SD, Pyle AD, Byrne JA, and Lipshutz GS
- Abstract
Urea cycle disorders are incurable enzymopathies that affect nitrogen metabolism and typically lead to hyperammonemia. Arginase deficiency results from a mutation in Arg1, the enzyme regulating the final step of ureagenesis and typically results in developmental disabilities, seizures, spastic diplegia, and sometimes death. Current medical treatments for urea cycle disorders are only marginally effective, and for proximal disorders, liver transplantation is effective but limited by graft availability. Advances in human induced pluripotent stem cell research has allowed for the genetic modification of stem cells for potential cellular replacement therapies. In this study, we demonstrate a universally-applicable CRISPR/Cas9-based strategy utilizing exon 1 of the hypoxanthine-guanine phosphoribosyltransferase locus to genetically modify and restore arginase activity, and thus ureagenesis, in genetically distinct patient-specific human induced pluripotent stem cells and hepatocyte-like derivatives. Successful strategies restoring gene function in patient-specific human induced pluripotent stem cells may advance applications of genetically modified cell therapy to treat urea cycle and other inborn errors of metabolism.
- Published
- 2016
- Full Text
- View/download PDF
33. Exon Skipping Therapy.
- Author
-
Young CS and Pyle AD
- Subjects
- Exons, Humans, Reading Frames, United States, United States Food and Drug Administration, Dystrophin genetics, Genetic Therapy, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne therapy
- Abstract
Exondys 51 is the first therapy for Duchenne muscular dystrophy (DMD) to have been granted accelerated approval by the FDA. Approval was granted based on using dystrophin expression as a surrogate marker. Exondys 51 targets DMD exon 51 for skipping to restore the reading frame for 13% of Duchenne patients., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
34. A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC-Derived Muscle Cells.
- Author
-
Young CS, Hicks MR, Ermolova NV, Nakano H, Jan M, Younesi S, Karumbayaram S, Kumagai-Cresse C, Wang D, Zack JA, Kohn DB, Nakano A, Nelson SF, Miceli MC, Spencer MJ, and Pyle AD
- Subjects
- Animals, Dystrophin deficiency, Dystrophin genetics, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells pathology, Mice, Mice, SCID, Muscle, Skeletal cytology, Muscle, Skeletal pathology, Muscular Dystrophy, Duchenne metabolism, Muscular Dystrophy, Duchenne pathology, CRISPR-Cas Systems genetics, Dystrophin metabolism, Gene Deletion, Gene Editing methods, Induced Pluripotent Stem Cells metabolism, Muscle, Skeletal metabolism, Muscular Dystrophy, Duchenne genetics
- Abstract
Mutations in DMD disrupt the reading frame, prevent dystrophin translation, and cause Duchenne muscular dystrophy (DMD). Here we describe a CRISPR/Cas9 platform applicable to 60% of DMD patient mutations. We applied the platform to DMD-derived hiPSCs where successful deletion and non-homologous end joining of up to 725 kb reframed the DMD gene. This is the largest CRISPR/Cas9-mediated deletion shown to date in DMD. Use of hiPSCs allowed evaluation of dystrophin in disease-relevant cell types. Cardiomyocytes and skeletal muscle myotubes derived from reframed hiPSC clonal lines had restored dystrophin protein. The internally deleted dystrophin was functional as demonstrated by improved membrane integrity and restoration of the dystrophin glycoprotein complex in vitro and in vivo. Furthermore, miR31 was reduced upon reframing, similar to observations in Becker muscular dystrophy. This work demonstrates the feasibility of using a single CRISPR pair to correct the reading frame for the majority of DMD patients., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. REST/NRSF Knockdown Alters Survival, Lineage Differentiation and Signaling in Human Embryonic Stem Cells.
- Author
-
Thakore-Shah K, Koleilat T, Jan M, John A, and Pyle AD
- Subjects
- Cell Line, Cell Survival, Humans, Repressor Proteins genetics, Cell Differentiation, Gene Knockdown Techniques, Human Embryonic Stem Cells metabolism, MAP Kinase Signaling System, Repressor Proteins metabolism
- Abstract
REST (RE1 silencing transcription factor), also known as NRSF (neuron-restrictive silencer factor), is a well-known transcriptional repressor of neural genes in non-neural tissues and stem cells. Dysregulation of REST activity is thought to play a role in diverse diseases including epilepsy, cancer, Down's syndrome and Huntington's disease. The role of REST/NRSF in control of human embryonic stem cell (hESC) fate has never been examined. To evaluate the role of REST in hESCs we developed an inducible REST knockdown system and examined both growth and differentiation over short and long term culture. Interestingly, we have found that altering REST levels in multiple hESC lines does not result in loss of self-renewal but instead leads to increased survival. During differentiation, REST knockdown resulted in increased MAPK/ERK and WNT signaling and increased expression of mesendoderm differentiation markers. Therefore we have uncovered a new role for REST in regulation of growth and early differentiation decisions in human embryonic stem cells.
- Published
- 2015
- Full Text
- View/download PDF
36. Molecular recognition enables nanosubstrate-mediated delivery of gene-encapsulated nanoparticles with high efficiency.
- Author
-
Peng J, Garcia MA, Choi JS, Zhao L, Chen KJ, Bernstein JR, Peyda P, Hsiao YS, Liu KW, Lin WY, Pyle AD, Wang H, Hou S, and Tseng HR
- Subjects
- Amino Acid Motifs, Animals, Female, Genetic Vectors, HEK293 Cells, HeLa Cells, Humans, Jurkat Cells, Light, MCF-7 Cells, Mice, Mice, Inbred BALB C, Microscopy, Electron, Scanning, Microscopy, Electron, Transmission, Microscopy, Fluorescence, NIH 3T3 Cells, Particle Size, Scattering, Radiation, beta-Cyclodextrins chemistry, Gene Transfer Techniques, Nanoparticles chemistry, Nanotechnology methods
- Abstract
Substrate-mediated gene delivery is a promising method due to its unique ability to preconcentrate exogenous genes onto designated substrates. However, many challenges remain to enable continuous and multiround delivery of the gene using the same substrates without depositing payloads and immobilizing cells in each round of delivery. Herein we introduce a gene delivery system, nanosubstrate-mediated delivery (NSMD) platform, based on two functional components with nanoscale features, including (1) DNA⊂SNPs, supramolecular nanoparticle (SNP) vectors for gene encapsulation, and (2) Ad-SiNWS, adamantane (Ad)-grafted silicon nanowire substrates. The multivalent molecular recognition between the Ad motifs on Ad-SiNWS and the β-cyclodextrin (CD) motifs on DNA⊂SNPs leads to dynamic assembly and local enrichment of DNA⊂SNPs from the surrounding medium onto Ad-SiNWS. Subsequently, once cells settled on the substrate, DNA⊂SNPs enriched on Ad-SiNWS were introduced through the cell membranes by intimate contact with individual nanowires on Ad-SiNWS, resulting in a highly efficient delivery of exogenous genes. Most importantly, sequential delivery of multiple batches of exogenous genes on the same batch cells settled on Ad-SiNWS was realized by sequential additions of the corresponding DNA⊂SNPs with equivalent efficiency. Moreover, using the NSMD platform in vivo, cells recruited on subcutaneously transplanted Ad-SiNWS were also efficiently transfected with exogenous genes loaded into SNPs, validating the in vivo feasibility of this system. We believe that this nanosubstrate-mediated delivery platform will provide a superior system for in vitro and in vivo gene delivery and can be further used for the encapsulation and delivery of other biomolecules.
- Published
- 2014
- Full Text
- View/download PDF
37. Generation and characterization of transgene-free human induced pluripotent stem cells and conversion to putative clinical-grade status.
- Author
-
Awe JP, Lee PC, Ramathal C, Vega-Crespo A, Durruthy-Durruthy J, Cooper A, Karumbayaram S, Lowry WE, Clark AT, Zack JA, Sebastiano V, Kohn DB, Pyle AD, Martin MG, Lipshutz GS, Phelps PE, Pera RA, and Byrne JA
- Subjects
- Animals, Cell Differentiation, Cellular Reprogramming, Gene Expression, Genomics, Humans, Mice, Transgenes, Induced Pluripotent Stem Cells metabolism
- Abstract
Introduction: The reprogramming of a patient's somatic cells back into induced pluripotent stem cells (iPSCs) holds significant promise for future autologous cellular therapeutics. The continued presence of potentially oncogenic transgenic elements following reprogramming, however, represents a safety concern that should be addressed prior to clinical applications. The polycistronic stem cell cassette (STEMCCA), an excisable lentiviral reprogramming vector, provides, in our hands, the most consistent reprogramming approach that addresses this safety concern. Nevertheless, most viral integrations occur in genes, and exactly how the integration, epigenetic reprogramming, and excision of the STEMCCA reprogramming vector influences those genes and whether these cells still have clinical potential are not yet known., Methods: In this study, we used both microarray and sensitive real-time PCR to investigate gene expression changes following both intron-based reprogramming and excision of the STEMCCA cassette during the generation of human iPSCs from adult human dermal fibroblasts. Integration site analysis was conducted using nonrestrictive linear amplification PCR. Transgene-free iPSCs were fully characterized via immunocytochemistry, karyotyping and teratoma formation, and current protocols were implemented for guided differentiation. We also utilized current good manufacturing practice guidelines and manufacturing facilities for conversion of our iPSCs into putative clinical grade conditions., Results: We found that a STEMCCA-derived iPSC line that contains a single integration, found to be located in an intronic location in an actively transcribed gene, PRPF39, displays significantly increased expression when compared with post-excised stem cells. STEMCCA excision via Cre recombinase returned basal expression levels of PRPF39. These cells were also shown to have proper splicing patterns and PRPF39 gene sequences. We also fully characterized the post-excision iPSCs, differentiated them into multiple clinically relevant cell types (including oligodendrocytes, hepatocytes, and cardiomyocytes), and converted them to putative clinical-grade conditions using the same approach previously approved by the US Food and Drug Administration for the conversion of human embryonic stem cells from research-grade to clinical-grade status., Conclusion: For the first time, these studies provide a proof-of-principle for the generation of fully characterized transgene-free human iPSCs and, in light of the limited availability of current good manufacturing practice cellular manufacturing facilities, highlight an attractive potential mechanism for converting research-grade cell lines into putatively clinical-grade biologics for personalized cellular therapeutics.
- Published
- 2013
- Full Text
- View/download PDF
38. Involvement of lysosomal exocytosis in the excretion of mesoporous silica nanoparticles and enhancement of the drug delivery effect by exocytosis inhibition.
- Author
-
Yanes RE, Tarn D, Hwang AA, Ferris DP, Sherman SP, Thomas CR, Lu J, Pyle AD, Zink JI, and Tamanoi F
- Subjects
- Cell Line, Exocytosis physiology, Humans, Drug Carriers chemistry, Lysosomes chemistry, Nanoparticles chemistry, Silicon Dioxide chemistry
- Abstract
The exocytosis of phosphonate modified mesoporous silica nanoparticles (P-MSNs) is demonstrated and lysosomal exocytosis is identified as the mechanism responsible for this event. Regulation of P-MSN exocytosis can be achieved by inhibiting or accelerating lysosomal exocytosis. Slowing down P-MSN exocytosis enhances the drug delivery effect of CPT-loaded P-MSNs by improving cell killing., (Copyright © 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2013
- Full Text
- View/download PDF
39. Nipah virus envelope-pseudotyped lentiviruses efficiently target ephrinB2-positive stem cell populations in vitro and bypass the liver sink when administered in vivo.
- Author
-
Palomares K, Vigant F, Van Handel B, Pernet O, Chikere K, Hong P, Sherman SP, Patterson M, An DS, Lowry WE, Mikkola HK, Morizono K, Pyle AD, and Lee B
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Molecular Biology methods, Nipah Virus genetics, Transduction, Genetic, Ephrin-B2 metabolism, Genetic Vectors, Lentivirus genetics, Nipah Virus physiology, Receptors, Virus metabolism, Stem Cells virology, Virus Internalization
- Abstract
Sophisticated retargeting systems for lentiviral vectors have been developed in recent years. Most seek to suppress the viral envelope's natural tropism while modifying the receptor-binding domain such that its tropism is determined by the specificity of the engineered ligand-binding motif. Here we took advantage of the natural tropism of Nipah virus (NiV), whose attachment envelope glycoprotein has picomolar affinity for ephrinB2, a molecule proposed as a molecular marker of "stemness" (present on embryonic, hematopoietic, and neural stem cells) as well as being implicated in tumorigenesis of specific cancers. NiV entry requires both the fusion (F) and attachment (G) glycoproteins. Truncation of the NiV-F cytoplasmic tail (T5F) alone, combined with full-length NiV-G, resulted in optimal titers of NiV-pseudotyped particles (NiVpp) (∼10(6) IU/ml), even without ultracentrifugation. To further enhance the infectivity of NiVpp, we engineered a hyperfusogenic NiV-F protein lacking an N-linked glycosylation site (T5FΔN3). T5FΔN3/wt G particles exhibited enhanced infectivity on less permissive cell lines and efficiently targeted ephrinB2(+) cells even in a 1,000-fold excess of ephrinB2-negative cells, all without any loss of specificity, as entry was abrogated by soluble ephrinB2. NiVpp also transduced human embryonic, hematopoietic, and neural stem cell populations in an ephrinB2-dependent manner. Finally, intravenous administration of the luciferase reporter NiVpp-T5FΔN3/G to mice resulted in signals being detected in the spleen and lung but not in the liver. Bypassing the liver sink is a critical barrier for targeted gene therapy. The extraordinary specificity of NiV-G for ephrinB2 holds promise for targeting specific ephrinB2(+) populations in vivo or in vitro.
- Published
- 2013
- Full Text
- View/download PDF
40. Small molecule screening with laser cytometry can be used to identify pro-survival molecules in human embryonic stem cells.
- Author
-
Sherman SP and Pyle AD
- Subjects
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine analogs & derivatives, 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine pharmacology, Algorithms, Animals, Cell Survival drug effects, DNA metabolism, Drug Evaluation, Preclinical economics, Embryonic Stem Cells metabolism, Green Fluorescent Proteins metabolism, High-Throughput Screening Assays, Humans, Laser Scanning Cytometry economics, Mice, Octamer Transcription Factor-3 metabolism, Propidium metabolism, Protein Kinase Inhibitors pharmacology, Spectrometry, Fluorescence, Staining and Labeling, Time Factors, rho-Associated Kinases antagonists & inhibitors, Drug Evaluation, Preclinical methods, Embryonic Stem Cells cytology, Embryonic Stem Cells drug effects, Laser Scanning Cytometry methods, Small Molecule Libraries pharmacology
- Abstract
Differentiated cells from human embryonic stem cells (hESCs) provide an unlimited source of cells for use in regenerative medicine. The recent derivation of human induced pluripotent cells (hiPSCs) provides a potential supply of pluripotent cells that avoid immune rejection and could provide patient-tailored therapy. In addition, the use of pluripotent cells for drug screening could enable routine toxicity testing and evaluation of underlying disease mechanisms. However, prior to establishment of patient specific cells for cell therapy it is important to understand the basic regulation of cell fate decisions in hESCs. One critical issue that hinders the use of these cells is the fact that hESCs survive poorly upon dissociation, which limits genetic manipulation because of poor cloning efficiency of individual hESCs, and hampers production of large-scale culture of hESCs. To address the problems associated with poor growth in culture and our lack of understanding of what regulates hESC signaling, we successfully developed a screening platform that allows for large scale screening for small molecules that regulate survival. In this work we developed the first large scale platform for hESC screening using laser scanning cytometry and were able to validate this platform by identifying the pro-survival molecule HA-1077. These small molecules provide targets for both improving our basic understanding of hESC survival as well as a tool to improve our ability to expand and genetically manipulate hESCs for use in regenerative applications.
- Published
- 2013
- Full Text
- View/download PDF
41. A spatially and chemically defined platform for the uniform growth of human pluripotent stem cells.
- Author
-
Jonas SJ, Alva JA, Richardson W, Sherman SP, Galic Z, Pyle AD, and Dunn B
- Subjects
- Biocompatible Materials pharmacology, Cell Adhesion drug effects, Cell Culture Techniques, Cell Differentiation drug effects, Cell Proliferation drug effects, Cells, Cultured, Collagen chemistry, Dimethylpolysiloxanes chemistry, Drug Combinations, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Humans, Karyotyping, Laminin chemistry, Pluripotent Stem Cells metabolism, Proteoglycans chemistry, Silicon chemistry, Transcription Factors metabolism, Biocompatible Materials chemistry, Pluripotent Stem Cells cytology
- Abstract
We report the design of a chemically defined platform engineered for the culture of human pluripotent stem cells (hPSCs) that supports the long-term maintenance of self-renewing hPSC populations in a more uniform manner than standard culture systems. Microcontact printing (μCP) of alkanethiol self-assembled monolayers (SAMs) was used to spatially direct hPSC adherence. This technique not only establishes control over hPSC colony size and shape but also preserves genetic stability and provides unprecedented uniformity in the pluripotency of hPSC populations that is quantitatively assessed in the present study., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
42. Insights into skeletal muscle development and applications in regenerative medicine.
- Author
-
Tran T, Andersen R, Sherman SP, and Pyle AD
- Subjects
- Aging pathology, Aging physiology, Animals, Dogs, Embryoid Bodies cytology, Gene Expression Regulation, Developmental, Humans, Mice, MicroRNAs genetics, Muscle Development genetics, Muscular Dystrophy, Animal etiology, Muscular Dystrophy, Animal therapy, Myoblasts, Skeletal cytology, Myoblasts, Skeletal physiology, Pluripotent Stem Cells cytology, Pluripotent Stem Cells physiology, Pluripotent Stem Cells transplantation, Receptors, Notch physiology, Regeneration genetics, Regenerative Medicine methods, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle physiology, Signal Transduction, Stem Cell Niche, Wnt Signaling Pathway, Muscle Development physiology, Muscle, Skeletal growth & development, Muscle, Skeletal physiology, Regeneration physiology
- Abstract
Embryonic and postnatal development of skeletal muscle entails highly regulated processes whose complexity continues to be deconstructed. One key stage of development is the satellite cell, whose niche is composed of multiple cell types that eventually contribute to terminally differentiated myotubes. Understanding these developmental processes will ultimately facilitate treatments of myopathies such as Duchenne muscular dystrophy (DMD), a disease characterized by compromised cell membrane structure, resulting in severe muscle wasting. One theoretical approach is to use pluripotent stem cells in a therapeutic setting to help replace degenerated muscle tissue. This chapter discusses key myogenic developmental stages and their regulatory pathways; artificial myogenic induction in pluripotent stem cells; advantages and disadvantages of DMD animal models; and therapeutic approaches targeting DMD. Furthermore, skeletal muscle serves as an excellent paradigm for understanding general cell fate decisions throughout development., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
43. Phosphatase and tensin homolog regulates the pluripotent state and lineage fate choice in human embryonic stem cells.
- Author
-
Alva JA, Lee GE, Escobar EE, and Pyle AD
- Subjects
- Animals, Cell Line, Cell Lineage, Cell Proliferation, Cell Survival, Embryonic Stem Cells metabolism, Gene Knockdown Techniques, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Lentivirus genetics, Lentivirus metabolism, Male, Mice, Mice, SCID, Nanog Homeobox Protein, Octamer Transcription Factor-3 genetics, Octamer Transcription Factor-3 metabolism, PTEN Phosphohydrolase genetics, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Pluripotent Stem Cells metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Signal Transduction, Teratoma genetics, Teratoma metabolism, Teratoma pathology, Cell Differentiation, Embryonic Stem Cells cytology, PTEN Phosphohydrolase metabolism, Pluripotent Stem Cells cytology
- Abstract
Understanding the intrinsic and extrinsic signals that regulate the molecular basis of the pluripotent state may improve our understanding of mammalian embryogenesis, different states of pluripotency, and our ability to tailor lineage differentiation. Although the role of the PI3K/Akt pathway in the self-renewal and maintenance of mESCs is well-established, the specific contribution of the pathway or of its negative regulator, PTEN, in the maintenance of the human pluripotent state is less understood. To explore the PI3K/AKT pathway in human embryonic stem cell (hESC) pluripotency and differentiation, we generated stable PTEN knockdown (KD) hESCs using short hairpin RNA. Similar to mESCs, we found that PTEN KD hESCs have increased self-renewal, cell survival, and proliferation over multiple passages compared to control cells. However, in contrast to mESCs, in vitro, PTEN KD hESCs differentiated inefficiently in directed differentiation assays, in part due to the continued maintenance of OCT4 and NANOG expression. In teratoma assays, PTEN KD hESCs generated tissues from the three germ layers, although with a bias toward neuroectoderm differentiation. These results demonstrate that PTEN is a key regulator of hESC growth and differentiation, and manipulation of this pathway may improve our ability to regulate and understand the pluripotent state., (Copyright © 2011 AlphaMed Press.)
- Published
- 2011
- Full Text
- View/download PDF
44. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner.
- Author
-
Le Belle JE, Orozco NM, Paucar AA, Saxe JP, Mottahedeh J, Pyle AD, Wu H, and Kornblum HI
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Mice, Inbred Strains, Neural Stem Cells metabolism, Phosphatidylinositol 3-Kinases genetics, Proto-Oncogene Proteins c-akt genetics, Cell Proliferation, Neural Stem Cells cytology, Neurogenesis physiology, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Reactive Oxygen Species metabolism, Signal Transduction
- Abstract
The majority of research on reactive oxygen species (ROS) has focused on their cellular toxicities. Stem cells generally have been thought to maintain low levels of ROS as a protection against these processes. However, recent studies suggest that ROS can also play roles as second messengers, activating normal cellular processes. Here, we investigated ROS function in primary brain-derived neural progenitors. Somewhat surprisingly, we found that proliferative, self-renewing multipotent neural progenitors with the phenotypic characteristics of neural stem cells (NSC) maintained a high ROS status and were highly responsive to ROS stimulation. ROS-mediated enhancements in self-renewal and neurogenesis were dependent on PI3K/Akt signaling. Pharmacological or genetic manipulations that diminished cellular ROS levels also interfered with normal NSC and/or multipotent progenitor function both in vitro and in vivo. This study has identified a redox-mediated regulatory mechanism of NSC function that may have significant implications for brain injury, disease, and repair., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. Human pluripotent stem cells: the development of high-content screening strategies.
- Author
-
Sherman SP, Alva JA, Thakore-Shah K, and Pyle AD
- Subjects
- Animals, Cell Proliferation, Cell Survival, Cells, Cultured, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Humans, Mice, Pluripotent Stem Cells metabolism, Reproducibility of Results, Software, Staining and Labeling, Statistics as Topic, Time Factors, High-Throughput Screening Assays methods, Pluripotent Stem Cells cytology
- Abstract
High-content screening (HCS) permits simultaneous observation and analysis of multiple cellular variables including cell morphology, survival, and differentiation in live cells at the single-cell level, at the level of the culture well, and across the entire culture. By combining high-throughput technologies such as robotics, chemical libraries, and automated high-resolution microscopy, scientists are able to evaluate a much broader array of experimental conditions than can be studied using conventional cell biological techniques that study fewer parameters at any one time. Thus, HCS assays provide a means to vastly improve our basic understanding of stem cell biology. We have developed a HCS assay that allows the study of the effects of hundreds of small molecules in parallel. The protocol described in this chapter was developed to assess the effects of small molecules on the survival, proliferation, and expression of pluripotent markers following single-cell dissociation of human embryonic stem cells, but can be applied to the study of other types of stem cells including induced pluripotent stem cells. A detailed protocol for the setup of HCS assays and the parameters used to identify chemical modifiers of survival in human pluripotent stem cells, as well as secondary assays used to validate the small-molecule "hits" obtained during the high-content screen, are described.
- Published
- 2011
- Full Text
- View/download PDF
46. Female human iPSCs retain an inactive X chromosome.
- Author
-
Tchieu J, Kuoy E, Chin MH, Trinh H, Patterson M, Sherman SP, Aimiuwu O, Lindgren A, Hakimian S, Zack JA, Clark AT, Pyle AD, Lowry WE, and Plath K
- Subjects
- Cell Culture Techniques, Cell Differentiation, Epigenesis, Genetic, Female, Fibroblasts cytology, Humans, Cellular Reprogramming, Induced Pluripotent Stem Cells, X Chromosome Inactivation
- Abstract
Generating induced pluripotent stem cells (iPSCs) requires massive epigenome reorganization. It is unclear whether reprogramming of female human cells reactivates the inactive X chromosome (Xi), as in mouse. Here we establish that human (h)iPSCs derived from several female fibroblasts under standard culture conditions carry an Xi. Despite the lack of reactivation, the Xi undergoes defined chromatin changes, and expansion of hiPSCs can lead to partial loss of XIST RNA. These results indicate that hiPSCs are epigenetically dynamic and do not display a pristine state of X inactivation with two active Xs as found in some female human embryonic stem cell lines. Furthermore, whereas fibroblasts are mosaic for the Xi, hiPSCs are clonal. This nonrandom pattern of X chromosome inactivation in female hiPSCs, which is maintained upon differentiation, has critical implications for clinical applications and disease modeling, and could be exploited for a unique form of gene therapy for X-linked diseases., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
47. Increased lysis of stem cells but not their differentiated cells by natural killer cells; de-differentiation or reprogramming activates NK cells.
- Author
-
Tseng HC, Arasteh A, Paranjpe A, Teruel A, Yang W, Behel A, Alva JA, Walter G, Head C, Ishikawa TO, Herschman HR, Cacalano N, Pyle AD, Park NH, and Jewett A
- Subjects
- Animals, Blotting, Western, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Humans, Interferon-gamma metabolism, Interleukin-2 pharmacology, Interleukin-6 metabolism, Interleukin-8 metabolism, Killer Cells, Natural drug effects, Mouth Neoplasms pathology, Neoplasms, Squamous Cell pathology, Neoplastic Stem Cells immunology, Stem Cells cytology, Stem Cells metabolism, Killer Cells, Natural immunology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology
- Abstract
The aims of this study are to demonstrate the increased lysis of stem cells but not their differentiated counterparts by the NK cells and to determine whether disturbance in cell differentiation is a cause for increased sensitivity to NK cell mediated cytotoxicity. Increased cytotoxicity and augmented secretion of IFN-gamma were both observed when PBMCs or NK cells were co-incubated with primary UCLA oral squamous carcinoma stem cells (UCLA-OSCSCs) when compared to differentiated UCLA oral squamous carcinoma cells (UCLA-OSCCs). In addition, human embryonic stem cells (hESCs) were also lysed greatly by the NK cells. Moreover, NK cells were found to lyse human Mesenchymal Stem Cells (hMSCs), human dental pulp stem cells (hDPSCs) and human induced pluripotent stem cells (hiPSCs) significantly more than their differentiated counterparts or parental lines from which they were derived. It was also found that inhibition of differentiation or reversion of cells to a less-differentiated phenotype by blocking NFkappaB or targeted knock down of COX2 in monocytes significantly augmented NK cell cytotoxicity and secretion of IFN-gamma. Taken together, these results suggest that stem cells are significant targets of the NK cell cytotoxicity. However, to support differentiation of a subset of tumor or healthy untransformed primary stem cells, NK cells may be required to lyse a number of stem cells and/or those which are either defective or incapable of full differentiation in order to lose their cytotoxic function and gain the ability to secrete cytokines (split anergy). Therefore, patients with cancer may benefit from repeated allogeneic NK cell transplantation for specific elimination of cancer stem cells.
- Published
- 2010
- Full Text
- View/download PDF
48. Microfluidic image cytometry for quantitative single-cell profiling of human pluripotent stem cells in chemically defined conditions.
- Author
-
Kamei K, Ohashi M, Gschweng E, Ho Q, Suh J, Tang J, For Yu ZT, Clark AT, Pyle AD, Teitell MA, Lee KB, Witte ON, and Tseng HR
- Subjects
- Cells, Cultured, Equipment Design, Equipment Failure Analysis, Humans, Reproducibility of Results, Sensitivity and Specificity, Cell Culture Techniques instrumentation, Cell Separation instrumentation, Flow Cytometry instrumentation, Gene Expression Profiling instrumentation, Microfluidic Analytical Techniques instrumentation, Microscopy instrumentation, Pluripotent Stem Cells cytology
- Abstract
Microfluidic image cytometry (MIC) has been developed to study phenotypes of various hPSC lines by screening several chemically defined serum/feeder-free conditions. A chemically defined hPSC culture was established using 20 ng mL(-1) of bFGF on 20 microg mL(-1) of Matrigel to grow hPSCs over a week in an undifferentiated state. Following hPSC culture, we conducted quantitative MIC to perform a single cell profiling of simultaneously detected protein expression (OCT4 and SSEA1). Using clustering analysis, we were able to systematically compare the characteristics of various hPSC lines in different conditions.
- Published
- 2010
- Full Text
- View/download PDF
49. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures.
- Author
-
Chin MH, Mason MJ, Xie W, Volinia S, Singer M, Peterson C, Ambartsumyan G, Aimiuwu O, Richter L, Zhang J, Khvorostov I, Ott V, Grunstein M, Lavon N, Benvenisty N, Croce CM, Clark AT, Baxter T, Pyle AD, Teitell MA, Pelegrini M, Plath K, and Lowry WE
- Subjects
- Animals, Cell Line, DNA Methylation, Embryonic Stem Cells cytology, Gene Expression Profiling, Genomic Instability, Histones genetics, Humans, Mice, MicroRNAs metabolism, Pluripotent Stem Cells cytology, Promoter Regions, Genetic, Embryonic Stem Cells metabolism, Gene Expression, Pluripotent Stem Cells metabolism
- Abstract
Induced pluripotent stem cells (iPSCs) outwardly appear to be indistinguishable from embryonic stem cells (ESCs). A study of gene expression profiles of mouse and human ESCs and iPSCs suggests that, while iPSCs are quite similar to their embryonic counterparts, a recurrent gene expression signature appears in iPSCs regardless of their origin or the method by which they were generated. Upon extended culture, hiPSCs adopt a gene expression profile more similar to hESCs; however, they still retain a gene expression signature unique from hESCs that extends to miRNA expression. Genome-wide data suggested that the iPSC signature gene expression differences are due to differential promoter binding by the reprogramming factors. High-resolution array profiling demonstrated that there is no common specific subkaryotypic alteration that is required for reprogramming and that reprogramming does not lead to genomic instability. Together, these data suggest that iPSCs should be considered a unique subtype of pluripotent cell.
- Published
- 2009
- Full Text
- View/download PDF
50. Integrated chemical genomics reveals modifiers of survival in human embryonic stem cells.
- Author
-
Damoiseaux R, Sherman SP, Alva JA, Peterson C, and Pyle AD
- Subjects
- Blotting, Western, Cell Line, Cell Survival genetics, Cell Survival physiology, Chromatin Immunoprecipitation, Flow Cytometry, Gene Expression Profiling, Humans, Karyotyping, Protein Kinase C metabolism, RNA, Small Interfering, rho-Associated Kinases metabolism, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Genomics methods
- Abstract
Understanding how survival is regulated in human embryonic stem cells (hESCs) could improve expansion of stem cells for production of cells for regenerative therapy. There is great variability in comparing the differentiation potential of multiple hESC lines. One reason for this is poor survival upon dissociation, which limits selection of homogeneous populations of cells. Understanding the complexity of survival signals has been hindered by the lack of a reproducible system to identify modulators of survival in pluripotent cells. We therefore developed a high-content screening approach with small molecules to examine hESC survival. We have identified novel small molecules that improve survival by inhibiting either Rho-kinase or protein kinase C. Importantly, small molecule targets were verified using short hairpin RNA. Rescreening with stable hESCs that were genetically altered to have increased survival enabled us to identify groups of pathway targets that are important for modifying survival. Understanding how survival is regulated in hESCs could overcome severe technical difficulties in the field, namely expansion of stem cells to improve production of cells and tissues for regenerative therapy.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.