Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cell Res Ther ; 15(1): 106, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627844

ABSTRACT

BACKGROUND: Although oncogenic RAS mutants are thought to exert mutagenic effects upon blood cells, it remains uncertain how a single oncogenic RAS impacts non-transformed multipotent hematopoietic stem or progenitor cells (HPCs). Such potential pre-malignant status may characterize HPCs in patients with RAS-associated autoimmune lymphoproliferative syndrome-like disease (RALD). This study sought to elucidate the biological and molecular alterations in human HPCs carrying monoallelic mutant KRAS (G13C) with no other oncogene mutations. METHODS: We utilized induced pluripotent stem cells (iPSCs) derived from two unrelated RALD patients. Isogenic HPC pairs harboring either wild-type KRAS or monoallelic KRAS (G13C) alone obtained following differentiation enabled reliable comparative analyses. The compound screening was conducted with an established platform using KRAS (G13C) iPSCs and differentiated HPCs. RESULTS: Cell culture assays revealed that monoallelic KRAS (G13C) impacted both myeloid differentiation and expansion characteristics of iPSC-derived HPCs. Comprehensive RNA-sequencing analysis depicted close clustering of HPC samples within the isogenic group, warranting that comparative studies should be performed within the same genetic background. When compared with no stimulation, iPSC-derived KRAS (G13C)-HPCs showed marked similarity with the wild-type isogenic control in transcriptomic profiles. After stimulation with cytokines, however, KRAS (G13C)-HPCs exhibited obvious aberrant cell-cycle and apoptosis responses, compatible with "dysregulated expansion," demonstrated by molecular and biological assessment. Increased BCL-xL expression was identified amongst other molecular changes unique to mutant HPCs. With screening platforms established for therapeutic intervention, we observed selective activity against KRAS (G13C)-HPC expansion in several candidate compounds, most notably in a MEK- and a BCL-2/BCL-xL-inhibitor. These two compounds demonstrated selective inhibitory effects on KRAS (G13C)-HPCs even with primary patient samples when combined. CONCLUSIONS: Our findings indicate that a monoallelic oncogenic KRAS can confer dysregulated expansion characteristics to non-transformed HPCs, which may constitute a pathological condition in RALD hematopoiesis. The use of iPSC-based screening platforms will lead to discovering treatments that enable selective inhibition of RAS-mutated HPC clones.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
2.
Autophagy ; 19(1): 224-240, 2023 01.
Article in English | MEDLINE | ID: mdl-35503435

ABSTRACT

Dietary restriction (DR) is known to promote autophagy to exert its longevity effect. While SAMS-1 (S-adenosyl methionine synthetase-1) has been shown to be a key mediator of the DR response, little is known about the roles of S-adenosyl methionine (SAM) and SAM-dependent methyltransferase in autophagy and DR-induced longevity. In this study, we show that DR and SAMS-1 repress the activity of SET-2, a histone H3K4 methyltransferase, by limiting the availability of SAM. Consequently, the reduced H3K4me3 levels promote the expression and activity of two transcription factors, HLH-30/TFEB and PHA-4/FOXA, which both regulate the transcription of autophagy-related genes. We then find that HLH-30/TFEB and PHA-4/FOXA act collaboratively on their common target genes to mediate the transcriptional response of autophagy-related genes and consequently the lifespan of the animals. Our study thus shows that the SAMS-1-SET-2 axis serves as a nutrient-sensing module to epigenetically coordinate the activation of HLH-30/TFEB and PHA-4/FOXA transcription factors to control macroautophagy/autophagy and longevity in response to DR.Abbreviations: ChIP: chromatin immunoprecipitation; ChIP-seq: chromatin immuno precipitation-sequencing; COMPASS: complex of proteins associated with Set1; DR: dietary restriction; GO: gene ontology; SAM: S-adenosyl methionine; SAMS-1: S-adenosyl methionine synthetase-1; TSS: transcription start site; WT: wild-type.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Animals , Longevity/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Histones/metabolism , Methylation , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Autophagy/genetics , Transcription Factors/metabolism , Methionine , Basic Helix-Loop-Helix Transcription Factors/metabolism
3.
J Biomed Sci ; 27(1): 29, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31987051

ABSTRACT

Currently there are no therapies for treating Alzheimer's disease (AD) that can effectively halt disease progression. Existing drugs such as acetylcholinesterase inhibitors or NMDA receptor antagonists offers only symptomatic benefit. More recently, transplantation of neural stem cells (NSCs) to treat neurodegenerative diseases, including AD, has been investigated as a new therapeutic approach. Transplanted cells have the potential to replace damaged neural circuitry and secrete neurotrophic factors to counter symptomatic deterioration or to alter lesion protein levels. However, since there are animal models that can recapitulate AD in its entirety, it is challenging to precisely characterize the positive effects of transplanting NSCs. In the present review, we discuss the types of mouse modeling system that are available and the effect in each model after human-derived NSC (hNSC) or murine-derived NSC (mNSC) transplantation. Taken together, results from studies involving NSC transplantation in AD models indicate that this strategy could serve as a new therapeutic approach.


Subject(s)
Alzheimer Disease/therapy , Neural Stem Cells/transplantation , Stem Cell Transplantation , Animals , Disease Models, Animal , Humans , Mice
4.
Stem Cell Res Ther ; 10(1): 185, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31234949

ABSTRACT

BACKGROUND: Disease modeling with patient-derived induced pluripotent stem cells (iPSCs) is a powerful tool for elucidating the mechanisms underlying disease pathogenesis and developing safe and effective treatments. Patient peripheral blood (PB) cells are used for iPSC generation in many cases since they can be collected with minimum invasiveness. To derive iPSCs that lack immunoreceptor gene rearrangements, hematopoietic stem and progenitor cells (HSPCs) are often targeted as the reprogramming source. However, the current protocols generally require HSPC mobilization and/or ex vivo expansion owing to their sparsity at the steady state and low reprogramming efficiencies, making the overall procedure costly, laborious, and time-consuming. METHODS: We have established a highly efficient method for generating iPSCs from non-mobilized PB-derived CD34+ HSPCs. The source PB mononuclear cells were obtained from 1 healthy donor and 15 patients and were kept frozen until the scheduled iPSC generation. CD34+ HSPC enrichment was done using immunomagnetic beads, with no ex vivo expansion culture. To reprogram the CD34+-rich cells to pluripotency, the Sendai virus vector SeVdp-302L was used to transfer four transcription factors: KLF4, OCT4, SOX2, and c-MYC. In this iPSC generation series, the reprogramming efficiencies, success rates of iPSC line establishment, and progression time were recorded. After generating the iPSC frozen stocks, the cell recovery and their residual transgenes, karyotypes, T cell receptor gene rearrangement, pluripotency markers, and differentiation capability were examined. RESULTS: We succeeded in establishing 223 iPSC lines with high reprogramming efficiencies from 15 patients with 8 different disease types. Our method allowed the rapid appearance of primary colonies (~ 8 days), all of which were expandable under feeder-free conditions, enabling robust establishment steps with less workload. After thawing, the established iPSC lines were verified to be pluripotency marker-positive and of non-T cell origin. A majority of the iPSC lines were confirmed to be transgene-free, with normal karyotypes. Their trilineage differentiation capability was also verified in a defined in vitro assay. CONCLUSION: This robust and highly efficient method enables the rapid and cost-effective establishment of transgene-free iPSC lines from a small volume of PB, thus facilitating the biobanking of patient-derived iPSCs and their use for the modeling of various diseases.


Subject(s)
Antigens, CD34/metabolism , Cellular Reprogramming/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Sendai virus/genetics , Adolescent , Adult , Aged , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cellular Reprogramming/genetics , Female , Flow Cytometry , Humans , Karyotyping , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Middle Aged , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Young Adult
5.
Stem Cell Reports ; 11(2): 380-394, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29983389

ABSTRACT

Oncogenic KRAS mutations in hematopoietic stem cells cause RAS-associated autoimmune lymphoproliferative syndrome-like disease (RALD). KRAS plays essential roles in stemness maintenance in some types of stem cells. However, its roles in pluripotent stem cells (PSCs) are poorly understood. Here, we investigated the roles of KRAS on stemness in the context of induced PSCs (iPSCs). We used KRAS mutant (G13C/WT) and wild-type isogenic (WT/WT) iPSCs from the same RALD patients, as well as wild-type (WTed/WT) and heterozygous knockout (Δed/WT) iPSCs, both obtained by genome editing from the same G13C/WT clone. Compared with WT iPSCs, G13C/WT iPSCs displayed enforced retention of self-renewal and suppressed capacity for neuronal differentiation, while Δed/WT iPSCs showed normalized cellular characteristics similar to those of isogenic WTed/WT cells. The KRAS-ERK pathway, but not the KRAS-PI3K pathway, was shown to govern these G13C/WT-specific phenotypes, indicating the strong impact of the KRAS-ERK signaling upon self-renewal and differentiation propensity in human iPSCs.


Subject(s)
Cell Differentiation , Cell Self Renewal , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Alleles , Autoimmune Lymphoproliferative Syndrome , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cells, Cultured , Chromosome Aberrations , DNA Mutational Analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Editing , Gene Expression Profiling , Genotype , Humans , Induced Pluripotent Stem Cells/drug effects , Karyotype , Molecular Imaging , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects
6.
Hum Gene Ther Methods ; 27(5): 197-208, 2016 10.
Article in English | MEDLINE | ID: mdl-27763786

ABSTRACT

Stable gene transfer into target cell populations via integrating viral vectors is widely used in stem cell gene therapy (SCGT). Accurate vector copy number (VCN) estimation has become increasingly important. However, existing methods of estimation such as real-time quantitative PCR are more restricted in practicality, especially during clinical trials, given the limited availability of sample materials from patients. This study demonstrates the application of an emerging technology called droplet digital PCR (ddPCR) in estimating VCN states in the context of SCGT. Induced pluripotent stem cells (iPSCs) derived from a patient with X-linked chronic granulomatous disease were used as clonable target cells for transduction with alpharetroviral vectors harboring codon-optimized CYBB cDNA. Precise primer-probe design followed by multiplex analysis conferred assay specificity. Accurate estimation of per-cell VCN values was possible without reliance on a reference standard curve. Sensitivity was high and the dynamic range of detection was wide. Assay reliability was validated by observation of consistent, reproducible, and distinct VCN clustering patterns for clones of transduced iPSCs with varying numbers of transgene copies. Taken together, use of ddPCR appears to offer a practical and robust approach to VCN estimation with a wide range of clinical and research applications.


Subject(s)
DNA Copy Number Variations/genetics , Genetic Therapy , Genetic Vectors/genetics , Granulomatous Disease, Chronic/genetics , Induced Pluripotent Stem Cells/metabolism , DNA Primers/genetics , Genetic Vectors/isolation & purification , Granulomatous Disease, Chronic/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Stem Cell Transplantation , Virus Integration/genetics
7.
Mol Ther Methods Clin Dev ; 2: 15046, 2015.
Article in English | MEDLINE | ID: mdl-26682238

ABSTRACT

For the treatment of monogenetic hematological disorders, restoration of transgene expression in affected cell populations is generally considered to have beneficial effects. However, X-linked chronic granulomatous disease (XCGD) is unique since the appearance of functional neutrophils in the peripheral blood following hematopoietic stem cell gene therapy is transient only. One contributing factor could be the occurrence of detrimental effects secondary to ectopic gp91phox expression in neutrophils, which has not been formally demonstrated previously. This study uses iPSCs to model XCGD, which allows the process of differentiation to be studied intensely in vitro. Alpharetroviral vectors carrying a ubiquitous promoter were used to drive the "ectopic" expression of codon optimized gp91phox cDNA. In the mature fraction of neutrophils differentiated from transduced XCGD-iPSCs, cellular recovery in terms of gp91phox expression and reactive oxygen species production was abruptly lost before cells had fully differentiated. Most critically, ectopic gp91phox expression could be identified clearly in the developing fraction of the transduced groups, which appeared to correspond with reduced cell viability. It is possible that this impedes further differentiation of developing neutrophils. Therefore, affording cellular protection from the detrimental effects of ectopic gp91phox expression may improve XCGD clinical outcomes.

8.
Opt Express ; 22(26): 31646-53, 2014 Dec 29.
Article in English | MEDLINE | ID: mdl-25607135

ABSTRACT

We fabricated a phosphor-conversion white light emitting diode (PC-WLED) using a thin-film flip-chip GaN LED with a roughened u-GaN surface (TFFC-SR-LED) that emits blue light at 450 nm wavelength with a conformal phosphor coating that converts the blue light into yellow light. It was found that the TFFC-SR-LED with the thin-film substrate removal process and surface roughening exhibits a power enhancement of 16.1% when compared with the TFFC-LED without a sapphire substrate. When a TFFC-SR-LED with phosphors on a Cu-metal packaging-base (TFFC-SR-Cu-WLED) was operated at a forward-bias current of 350 mA, luminous flux and luminous efficacy were increased by 17.8 and 11.9%, compared to a TFFC-SR-LED on a Cup-shaped packaging-base (TFFC-SR-Cup-WLED). The angular correlated color temperature (CCT) deviation of a TFFC-SR-Cu-WLED reaches 77 K in the range of -70° to + 70° when the average CCT of white LEDs is around 4300 K. Consequently, the TFFC-SR-LED in a conformal coating phosphor structure on a Cu packaging-base could not only increase the luminous flux output, but also improve the angular-dependent CCT uniformity, thereby reducing the yellow ring effect.


Subject(s)
Color , Gallium/chemistry , Lasers , Lighting/instrumentation , Membranes, Artificial , Semiconductors , Equipment Design , Equipment Failure Analysis , Temperature
9.
Philos Trans R Soc Lond B Biol Sci ; 368(1609): 20110334, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23166396

ABSTRACT

In recent times, the epigenetic study of pluripotency based on cellular reprogramming techniques led to the creation of induced pluripotent stem cells. It has come to represent the forefront of a new wave of alternative therapeutic approaches in the field of stem cell therapy. Progress in drug development has saved countless lives, but there are numerous intractable diseases where curative treatment cannot be achieved through pharmacological intervention alone. Consequently, there has been an unfortunate rise in incidences of organ failures, degenerative disorders and cancers, hence novel therapeutic interventions are required. Stem cells have unique self-renewal and multilineage differentiation capabilities that could be harnessed for therapeutic purposes. Although a number of mature differentiated cells have been characterized in vitro, few have been demonstrated to function in a physiologically relevant context. Despite fervent levels of enthusiasm in the field, the reality is that other than the employment of haematopoietic stem cells, many other therapies have yet to be thoroughly proven for their therapeutic benefit and safety in application. This review shall focus on a discussion regarding the current status of stem cell therapy, the issues surrounding it and its future prospects with a general background on the regulatory networks underlying pluripotency.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Regeneration , Stem Cell Transplantation/methods , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Lineage , Central Nervous System Diseases/therapy , Diabetes Mellitus/therapy , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Pancreas/cytology , Pancreas/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...