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











Publication year range
1.
BMC Med Imaging ; 24(1): 218, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160500

ABSTRACT

Uterine fibroids are common benign tumors originating from the uterus's smooth muscle layer, often leading to symptoms such as pelvic pain, and reproductive issues. Early detection is crucial to prevent complications such as infertility or the need for invasive treatments like hysterectomy. One of the main challenges in diagnosing uterine fibroids is the lack of specific symptoms, which can mimic other gynecological conditions. This often leads to under-diagnosis or misdiagnosis, delaying appropriate management. In this research, an attention based fine-tuned EfficientNetB0 model is proposed for the classification of uterine fibroids from ultrasound images. Attention mechanisms, permit the model to focus on particular parts of an image and move forward the model's execution by empowering it to specifically go to imperative highlights whereas overlooking irrelevant ones. The proposed approach has used a total of 1990 images divided into two classes: Non-uterine fibroid and uterine fibroid. The data augmentation methods have been connected to improve generalization and strength by exposing it to a wider range of varieties within the training data. The proposed model has obtained the value of accuracy as 0.99. Future research should focus on improving the accuracy and efficiency of diagnostic techniques, as well as evaluating their effectiveness in diverse populations with higher sensitivity and specificity for the detection of uterine fibroids, as well as biomarkers to aid in diagnosis.


Subject(s)
Deep Learning , Leiomyoma , Ultrasonography , Uterine Neoplasms , Humans , Leiomyoma/diagnostic imaging , Female , Uterine Neoplasms/diagnostic imaging , Ultrasonography/methods , Sensitivity and Specificity , Image Interpretation, Computer-Assisted/methods
3.
Nat Cell Biol ; 25(12): 1758-1773, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919520

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.


Subject(s)
Muscle, Skeletal , PAX7 Transcription Factor , Regeneration , Satellite Cells, Skeletal Muscle , Animals , Humans , Mice , Muscle, Skeletal/physiology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Pluripotent Stem Cells , Satellite Cells, Skeletal Muscle/physiology
4.
iScience ; 25(11): 105415, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36388984

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.

5.
Mol Ther Nucleic Acids ; 29: 979-995, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36189080

ABSTRACT

The use of T cells from healthy donors for allogeneic chimeric antigen receptor T (CAR-T) cell cancer therapy is attractive because healthy donor T cells can produce versatile off-the-shelf CAR-T treatments. To maximize safety and durability of allogeneic products, the endogenous T cell receptor and major histocompatibility complex class I molecules are often removed via knockout of T cell receptor beta constant (TRBC) (or T cell receptor alpha constant [TRAC]) and B2M, respectively. However, gene editing tools (e.g., CRISPR-Cas9) can display poor fidelity, which may result in dangerous off-target mutations. Additionally, many gene editing technologies require T cell activation, resulting in a low percentage of desirable stem cell memory T cells (TSCM). We characterize an RNA-guided endonuclease, called Cas-CLOVER, consisting of the Clo051 nuclease domain fused with catalytically dead Cas9. In primary T cells from multiple donors, we find that Cas-CLOVER is a high-fidelity site-specific nuclease, with low off-target activity. Notably, Cas-CLOVER yields efficient multiplexed gene editing in resting T cells. In conjunction with the piggyBac transposon for delivery of a CAR transgene against the B cell maturation antigen (BCMA), we produce allogeneic CAR-T cells composed of high percentages of TSCM cells and possessing potent in vivo anti-tumor cytotoxicity.

6.
Nat Cancer ; 3(8): 961-975, 2022 08.
Article in English | MEDLINE | ID: mdl-35982179

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.


Subject(s)
Rhabdomyosarcoma, Embryonal , Rhabdomyosarcoma , Child , Humans , Muscle, Skeletal/pathology , Rhabdomyosarcoma/genetics , Single-Cell Analysis , Stem Cells/pathology
8.
Exp Cell Res ; 411(2): 112990, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34973262

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.


Subject(s)
Muscle Development/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , 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
9.
Nat Commun ; 12(1): 2595, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972536

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.


Subject(s)
Epidermis/anatomy & histology , Epidermis/metabolism , Hair Follicle/metabolism , Morphogenesis/physiology , Regeneration/physiology , Twist-Related Protein 1/metabolism , Wound Healing/physiology , 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
10.
STAR Protoc ; 1(3): 100158, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377052

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).


Subject(s)
Genes, Reporter , Muscle Development , PAX7 Transcription Factor/metabolism , Pluripotent Stem Cells/metabolism , 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, Kinetoplastida/genetics , Reproducibility of Results , Somites/embryology
12.
Cell Stem Cell ; 27(1): 158-176.e10, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32396864

ABSTRACT

The developmental trajectory of human skeletal myogenesis and the transition between progenitor and stem cell states are unclear. We used single-cell RNA sequencing to profile human skeletal muscle tissues from embryonic, fetal, and postnatal stages. In silico, we identified myogenic as well as other cell types and constructed a "roadmap" of human skeletal muscle ontogeny across development. In a similar fashion, we also profiled the heterogeneous cell cultures generated from multiple human pluripotent stem cell (hPSC) myogenic differentiation protocols and mapped hPSC-derived myogenic progenitors to an embryonic-to-fetal transition period. We found differentially enriched biological processes and discovered co-regulated gene networks and transcription factors present at distinct myogenic stages. This work serves as a resource for advancing our knowledge of human myogenesis. It also provides a tool for a better understanding of hPSC-derived myogenic progenitors for translational applications in skeletal muscle-based regenerative medicine.


Subject(s)
Muscle Development , Pluripotent Stem Cells , Cell Differentiation , Humans , Muscle, Skeletal , Transcription Factors
13.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31852842

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-ß+CD146+CD34-CD56- pericytes that lack expression of the fibro-adipogenic cell marker PDGFR-α. Accordingly, the PDGFR-ß+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-ß+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.


Subject(s)
Human Embryonic Stem Cells/physiology , Muscular Disorders, Atrophic/therapy , Pericytes/transplantation , Rotator Cuff Injuries/complications , Rotator Cuff/pathology , 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
14.
Science ; 366(6466): 684-685, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31699921
15.
Nat Cell Biol ; 20(1): 46-57, 2018 01.
Article in English | MEDLINE | ID: mdl-29255171

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-ß 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.


Subject(s)
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 , 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
16.
Cell Rep ; 18(6): 1573-1585, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28178531

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-ß-catenin, we also identified BMP and transforming growth factor ß (TGF-ß) 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.


Subject(s)
Embryonic Development/physiology , Morphogenesis/physiology , Pluripotent Stem Cells/physiology , Somites/physiology , 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
17.
IUBMB Life ; 66(2): 110-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24578297

ABSTRACT

Through the eons of time, out of all possible configurations, nature has selected glucose not only as a vital source of energy to sustain life but also as the molecule who's structure supplies the appropriate elements required for a cell to grow and multiply. This understanding, at least in part, explains the profound effects that the analog of glucose, 2-deoxy-d-glucose, has been shown to have on as common and widespread diseases as cancer, viral infection, aging-related morbidity, epilepsy, and others. This review is confined to summarizing some of the salient findings of this remarkable compound as they relate mainly to cancer.


Subject(s)
Deoxyglucose/metabolism , Endoplasmic Reticulum Stress/genetics , Neoplasms/metabolism , Virus Replication/genetics , Apoptosis/genetics , Autophagy/genetics , Deoxyglucose/genetics , Glycosylation , Humans , Hypoxia , Neoplasms/genetics , Neoplasms/pathology
18.
Cancer Chemother Pharmacol ; 72(1): 251-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23700291

ABSTRACT

BACKGROUND: Inhibition of glucose metabolism has recently become an attractive target for cancer treatment. Accordingly, since 2-deoxyglucose (2-DG) competes effectively with glucose, it has come under increasing scrutiny as a therapeutic agent. The initial response of tumor cells to 2-DG is growth inhibition, which is thought to conserve energy and consequently protect cells from its ATP-lowering effects as a glycolytic inhibitor. However, since 2-DG also mimics mannose and thereby interferes with N-linked glycosylation, the question is raised of how this sugar analog inhibits tumor cell growth and whether the mechanism by which it protects cells can be manipulated to convert 2-DG-induced growth inhibition to cell death. METHODS: Cell growth and death were measured via counting viable and dead cells based on trypan blue exclusion. Markers of ATP reduction and the unfolded protein response (UPR) were detected by Western blot. Protein functions were manipulated through chemical compounds, siRNA and the use of gene-specific wild-type and knock-out mouse embryonic fibroblasts (MEFs). RESULTS: At 2-DG concentrations that can be achieved in human plasma without causing significant side effects, we find (a) It induces growth inhibition predominantly by interference with glycosylation, which leads to accumulation of unfolded proteins in the endoplasmic reticulum activating the UPR; (b) Inhibition of PERK (but not ATF6 or IRE1), a major component of the UPR, leads to conversion of 2-DG-induced growth inhibition to cell death and (c) secondarily to PERK, inhibition of GCN2, a kinase that is activated in response to low intracellular glutamine, increases 2-DG's cytotoxic effects in PERK -/- MEFs. CONCLUSIONS: Overall, these findings present a novel anticancer strategy that can be translated into therapeutic gain as they uncover the metabolic target PERK, and to a lesser degree GCN2, that when inhibited convert 2-DG's static effect to a toxic one in tumor cells growing under normoxia.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxyglucose/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Unfolded Protein Response/drug effects , eIF-2 Kinase/antagonists & inhibitors , Activating Transcription Factor 6/antagonists & inhibitors , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Biomarkers/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Glycosylation/drug effects , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
19.
Biochem Pharmacol ; 85(10): 1463-77, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23500541

ABSTRACT

Autophagy, a well-conserved cellular self-eating process, has been shown to play a critical role in the pathophysiology of cancer. Previously, we reported that under normal O2 conditions (21% O2), the dual glucose metabolism inhibitor 2-deoxyglucose (2-DG) activates a cytoprotective autophagic response in cancer cells mainly through the induction of endoplasmic reticulum (ER) stress rather than ATP² reduction. However, the pathway(s) by which this occurs was unknown. Here, we find that ER stress induced by 2-DG as well as tunicamycin activates AMPK via Ca²âº-CaMKKß leading to stimulation of autophagy. These results suggest a new role for AMPK as a sensor of ER stress. In contrast, we find that although physiologic glucose starvation (GS) leads to ER stress which contributes to autophagy activation, it does so by a different mechanism. In addition to ER stress, GS also stimulates autophagy through lowering ATP and activating the canonical LKB1-AMPK energy sensing pathway as well as through increasing reactive oxygen species resulting in the activation of ERK. Furthermore, under hypoxia we observe that both 2-DG and GS inhibit rather than activate autophagy. This inhibition correlates with dramatically depleted ATP levels, and occurs through reduction of the PI3K III-Beclin1 complex for autophagy initiation, blockage of the conjugation of ATG12 to ATG5 for autophagosome expansion, as well as inhibition of the functional lysosomal compartment for autophagic degradation. Taken together, our data support a model where under normoxia therapeutic (2-DG) and physiologic (GS) glucose restriction differentially activate autophagy, while under hypoxia they similarly inhibit it.


Subject(s)
Autophagy/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Deoxyglucose/deficiency , Endoplasmic Reticulum Stress/drug effects , Glucose/deficiency , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Deoxyglucose/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species , Signal Transduction/drug effects , Tunicamycin/pharmacology
20.
Cancer Chemother Pharmacol ; 67(4): 899-910, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20593179

ABSTRACT

PURPOSE: The glucose analog and glycolytic inhibitor 2-deoxy-D-glucose (2-DG), which is currently under clinical evaluation for targeting cancer cells, not only blocks glycolysis thereby reducing cellular ATP, but also interferes with N-linked glycosylation, which leads to endoplasmic reticulum (ER) stress and an unfolded protein response (UPR). Both bioenergetic challenge and ER stress have been shown to activate autophagy, a bulk cellular degradation process that plays either a pro- or anti-death role. Here, we investigate which pathway 2-DG interferes with that activates autophagy and the role of this process in modulating 2-DG-induced toxicity. METHODS: Pancreatic cancer cell line 1420, melanoma cell line MDA-MB-435 and breast cancer cell line SKBR3 were used to investigate the relationship between induction by 2-DG treatment of ER stress/UPR, ATP reduction and activation of autophagy. ER stress/UPR (Grp78 and CHOP) and autophagy (LC3B II) markers were assayed by immunoblotting, while ATP levels were measured using the CellTiter-Glo Luminescent Cell Viability Assay. Autophagy was also measured by immunofluorescence utilizing LC3B antibody. Cell death was detected with a Vi-Cell cell viability analyzer using trypan blue exclusion. RESULTS: In the three different cancer cell lines described earlier, we find that 2-DG upregulates autophagy, increases ER stress and lowers ATP levels. Addition of exogenous mannose reverses 2-DG-induced autophagy and ER stress but does not recover the lowered levels of ATP. Moreover, under anaerobic conditions where 2-DG severely depletes ATP, autophagy is diminished rather than activated, which correlates with lowered levels of the ER stress marker Grp78. Additionally, when autophagy is blocked by siRNA, cell sensitivity to 2-DG is increased corresponding with upregulation of ER stress-mediated apoptosis. Similar increased toxicity is observed with 3-methyladenine, a known autophagy inhibitor. In contrast, rapamycin which enhances autophagy reduces 2-DG-induced toxicity. CONCLUSIONS: Overall, these results indicate that the major mechanism by which 2-DG stimulates autophagy is through ER stress/UPR and not by lowering ATP levels. Furthermore, autophagy plays a protective role against 2-DG-elicited cell death apparently by relieving ER stress. These data suggest that combining autophagy inhibitors with 2-DG may be useful clinically.


Subject(s)
Antimetabolites/pharmacology , Autophagy/drug effects , Deoxyglucose/pharmacology , Endoplasmic Reticulum/drug effects , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Female , Fluorescent Antibody Technique , Humans , Melanoma/drug therapy , Melanoma/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Unfolded Protein Response/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL