Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
ACS Appl Bio Mater ; 6(9): 3790-3797, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37647213

ABSTRACT

There is an urgent need for simple and non-invasive identification of live neural stem/progenitor cells (NSPCs) in the developing and adult brain as well as in disease, such as in brain tumors, due to the potential clinical importance in prognosis, diagnosis, and treatment of diseases of the nervous system. Here, we report a luminescent conjugated oligothiophene (LCO), named p-HTMI, for non-invasive and non-amplified real-time detection of live human patient-derived glioblastoma (GBM) stem cell-like cells and NSPCs. While p-HTMI stained only a small fraction of other cell types investigated, the mere addition of p-HTMI to the cell culture resulted in efficient detection of NSPCs or GBM cells from rodents and humans within minutes. p-HTMI is functionalized with a methylated imidazole moiety resembling the side chain of histidine/histamine, and non-methylated analogues were not functional. Cell sorting experiments of human GBM cells demonstrated that p-HTMI labeled the same cell population as CD271, a proposed marker for stem cell-like cells and rapidly migrating cells in glioblastoma. Our results suggest that the LCO p-HTMI is a versatile tool for immediate and selective detection of neural and glioma stem and progenitor cells.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Stem Cells , Adult , Humans , Glioblastoma/diagnosis , Brain , Brain Neoplasms/diagnosis , Adapalene
2.
Cell Rep ; 28(8): 2064-2079.e11, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433983

ABSTRACT

Identifying cellular programs that drive cancers to be stem-like and treatment resistant is critical to improving outcomes in patients. Here, we demonstrate that constitutive extracellular signal-regulated kinase 1/2 (ERK1/2) activation sustains a stem-like state in glioblastoma (GBM), the most common primary malignant brain tumor. Pharmacological inhibition of ERK1/2 activation restores neurogenesis during murine astrocytoma formation, inducing neuronal differentiation in tumorspheres. Constitutive ERK1/2 activation globally regulates miRNA expression in murine and human GBMs, while neuronal differentiation of GBM tumorspheres following the inhibition of ERK1/2 activation requires the functional expression of miR-124 and the depletion of its target gene SOX9. Overexpression of miR124 depletes SOX9 in vivo and promotes a stem-like-to-neuronal transition, with reduced tumorigenicity and increased radiation sensitivity. Providing a rationale for reports demonstrating miR-124-induced abrogation of GBM aggressiveness, we conclude that reversal of an ERK1/2-miR-124-SOX9 axis induces a neuronal phenotype and that enforcing neuronal differentiation represents a therapeutic strategy to improve outcomes in GBM.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Glioblastoma/pathology , MAP Kinase Signaling System , MicroRNAs/metabolism , Neurons/pathology , SOX9 Transcription Factor/metabolism , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Benzamides/pharmacology , Brain Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Disease Progression , Female , Glioblastoma/genetics , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects
3.
Cell Stem Cell ; 25(3): 433-446.e7, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31204176

ABSTRACT

Human neural stem cell cultures provide progenitor cells that are potential cells of origin for brain cancers. However, the extent to which genetic predisposition to tumor formation can be faithfully captured in stem cell lines is uncertain. Here, we evaluated neuroepithelial stem (NES) cells, representative of cerebellar progenitors. We transduced NES cells with MYCN, observing medulloblastoma upon orthotopic implantation in mice. Significantly, transcriptomes and patterns of DNA methylation from xenograft tumors were globally more representative of human medulloblastoma compared to a MYCN-driven genetically engineered mouse model. Orthotopic transplantation of NES cells generated from Gorlin syndrome patients, who are predisposed to medulloblastoma due to germline-mutated PTCH1, also generated medulloblastoma. We engineered candidate cooperating mutations in Gorlin NES cells, with mutation of DDX3X or loss of GSE1 both accelerating tumorigenesis. These findings demonstrate that human NES cells provide a potent experimental resource for dissecting genetic causation in medulloblastoma.


Subject(s)
Basal Cell Nevus Syndrome/genetics , Brain Neoplasms/genetics , Medulloblastoma/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neural Stem Cells/physiology , Neuroepithelial Cells/physiology , Pluripotent Stem Cells/physiology , Animals , Basal Cell Nevus Syndrome/metabolism , Basal Cell Nevus Syndrome/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Genetic Engineering , Genetic Predisposition to Disease , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, SCID , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Proteins/genetics , Patched-1 Receptor/genetics , Stem Cell Transplantation , Transplantation, Heterologous
4.
Mol Cancer Res ; 16(5): 777-790, 2018 05.
Article in English | MEDLINE | ID: mdl-29431617

ABSTRACT

Interstitial fluid pressure (IFP) presents a barrier to drug uptake in solid tumors, including the aggressive primary brain tumor glioblastoma (GBM). It remains unclear how fluid dynamics impacts tumor progression and can be targeted therapeutically. To address this issue, a novel telemetry-based approach was developed to measure changes in IFP during progression of GBM xenografts. Antisecretory factor (AF) is an endogenous protein that displays antisecretory effects in animals and patients. Here, endogenous induction of AF protein or exogenous administration of AF peptide reduced IFP and increased drug uptake in GBM xenografts. AF inhibited cell volume regulation of GBM cells, an effect that was phenocopied in vitro by the sodium-potassium-chloride cotransporter 1 (SLC12A2/NKCC1) inhibitor bumetanide. As a result, AF induced apoptosis and increased survival in GBM models. In vitro, the ability of AF to reduce GBM cell proliferation was phenocopied by bumetanide and NKCC1 knockdown. Next, AF's ability to sensitize GBM cells to the alkylating agent temozolomide, standard of care in GBM patients, was evaluated. Importantly, combination of AF induction and temozolomide treatment blocked regrowth in GBM xenografts. Thus, AF-mediated inhibition of cell volume regulation represents a novel strategy to increase drug uptake and improve outcome in GBM. Mol Cancer Res; 16(5); 777-90. ©2018 AACR.


Subject(s)
Glioblastoma/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Cell Size , Disease Progression , Glioblastoma/pathology , Humans , Mice , Mice, Nude
5.
Cancer Cell ; 31(3): 424-435, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28292440

ABSTRACT

Although signaling from phosphatidylinositol 3-kinase (PI3K) and AKT to mechanistic target of rapamycin (mTOR) is prominently dysregulated in high-grade glial brain tumors, blockade of PI3K or AKT minimally affects downstream mTOR activity in glioma. Allosteric mTOR inhibitors, such as rapamycin, incompletely block mTORC1 compared with mTOR kinase inhibitors (TORKi). Here, we compared RapaLink-1, a TORKi linked to rapamycin, with earlier-generation mTOR inhibitors. Compared with rapamycin and Rapalink-1, TORKi showed poor durability. RapaLink-1 associated with FKBP12, an abundant mTOR-interacting protein, enabling accumulation of RapaLink-1. RapaLink-1 showed better efficacy than rapamycin or TORKi, potently blocking cancer-derived, activating mutants of mTOR. Our study re-establishes mTOR as a central target in glioma and traces the failure of existing drugs to incomplete/nondurable inhibition of mTORC1.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Multiprotein Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Sirolimus/therapeutic use , Tacrolimus Binding Protein 1A/physiology
6.
Sci Rep ; 6: 19727, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26812940

ABSTRACT

Neural crest cells (NCC) are stem cells that generate different lineages, including neuroendocrine, melanocytic, cartilage, and bone. The differentiation potential of NCC varies according to the level from which cells emerge along the neural tube. For example, only anterior "cranial" NCC form craniofacial bone, whereas solely posterior "trunk" NCC contribute to sympathoadrenal cells. Importantly, the isolation of human fetal NCC carries ethical and scientific challenges, as NCC induction typically occur before pregnancy is detectable. As a result, current knowledge of NCC biology derives primarily from non-human organisms. Important differences between human and non-human NCC, such as expression of HNK1 in human but not mouse NCC, suggest a need to study human NCC directly. Here, we demonstrate that current protocols to differentiate human pluripotent stem cells (PSC) to NCC are biased toward cranial NCC. Addition of retinoic acid drove trunk-related markers and HOX genes characteristic of a posterior identity. Subsequent treatment with bone morphogenetic proteins (BMPs) enhanced differentiation to sympathoadrenal cells. Our approach provides methodology for detailed studies of human NCC, and clarifies roles for retinoids and BMPs in the differentiation of human PSC to trunk NCC and to sympathoadrenal lineages.


Subject(s)
Cell Differentiation , Neural Crest/cytology , Neural Crest/embryology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Biomarkers , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Pluripotent Stem Cells/drug effects , Signal Transduction , Tretinoin/metabolism , Tretinoin/pharmacology
7.
Cancer Res ; 75(20): 4302-11, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26282165

ABSTRACT

High grade gliomas (HGG) are classified into four subgroups based on transcriptional signatures and phenotypic characteristics. In particular, the proneural-to-mesenchymal transition (PMT) is associated with increased malignancy, poor prognosis, and disease recurrence, but the underlying causes of PMT are still unclear. In this study, we investigated whether radiotherapy promotes PMT using a genetically engineered mouse model of proneural HGG. We found that cranial ionizing radiation induced robust and durable PMT in tumors. Additionally, we isolated primary proneural HGG cells from mouse and human tumors and demonstrate that radiation induced a sustained cell-intrinsic mesenchymal transition associated with increased invasiveness and resistance to the alkylating agent temozolomide. Expectedly, irradiation-induced PMT was also associated with activation of the STAT3 transcription factor, and the combination of STAT3 blockade using JAK2 inhibitors with radiation abrogated the mesenchymal transition and extended survival of mice. Taken together, our data suggest that clinical JAK2 inhibitors should be tested in conjunction with radiation in patients with proneural HGG as a new strategy for blocking the emergence of therapy-resistant mesenchymal tumors at relapse.


Subject(s)
Glioma/metabolism , Glioma/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Allografts , Animals , Biomarkers , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Knockout , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation , STAT3 Transcription Factor/metabolism
8.
Cell Rep ; 8(3): 665-70, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25088415

ABSTRACT

Bone morphogenetic proteins (BMPs) secreted by the dorsal neural tube and overlying ectoderm are key signals for the specification of the roof plate and dorsal interneuron populations. However, the signals that confer nonneurogenic character to the roof plate region are largely unknown. We report that the roof plate region shows elevated oxygen levels compared to neurogenic regions of the neural tube. These high oxygen levels are required for the expression of the antineuronal transcription factor Hes1 in the roof plate region. The transcriptional corepressor CtBP is a critical mediator of the oxygen-sensing response. High oxygen promotes a decrease in the CtBP occupancy of the promoter of Hes1. Furthermore, under conditions of high oxygen and BMP, CtBP associates with HES1 and represses neurogenesis. We propose that CtBP integrates signals originating from microenvironmental levels of oxygen and BMP to confer nonneurogenic character to the roof plate region.


Subject(s)
Carrier Proteins/metabolism , Eye Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Oxygen/metabolism , Stem Cell Niche , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/genetics , Cell Hypoxia , Cells, Cultured , Chick Embryo , Eye Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neural Stem Cells/cytology , Neural Tube/cytology , Neural Tube/metabolism , Promoter Regions, Genetic , Rats , Transcription Factor HES-1 , Transcription Factors/genetics
9.
Adv Cancer Res ; 121: 1-65, 2014.
Article in English | MEDLINE | ID: mdl-24889528

ABSTRACT

Glioma is the most common primary malignant brain tumor and arises throughout the central nervous system. Recent focus on stem-like glioma cells has implicated neural stem cells (NSCs), a minor precursor population restricted to germinal zones, as a potential source of gliomas. In this review, we focus on the relationship between oligodendrocyte progenitor cells (OPCs), the largest population of cycling glial progenitors in the postnatal brain, and gliomagenesis. OPCs can give rise to gliomas, with signaling pathways associated with NSCs also playing key roles during OPC lineage development. Gliomas can also undergo a switch from progenitor- to stem-like phenotype after therapy, consistent with an OPC-origin even for stem-like gliomas. Future in-depth studies of OPC biology may shed light on the etiology of OPC-derived gliomas and reveal new therapeutic avenues.


Subject(s)
Cell Transformation, Neoplastic , Glioma/pathology , Neural Stem Cells/physiology , Neuroglia/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/physiology , Cell Transformation, Neoplastic/pathology , Glioma/genetics , Glioma/therapy , Humans , Molecular Targeted Therapy , Neural Stem Cells/pathology , Neuroglia/pathology
11.
Biomaterials ; 30(27): 4567-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19500834

ABSTRACT

Microenvironmental mechanical properties of stem cell niches vary across tissues and developmental stages. Accumulating evidence suggests that matching substrate elasticity with in vivo tissue elasticity facilitates stem cell differentiation. However, it has not been established whether substrate elasticity can control the maturation stage of cells generated by stem cell differentiation. Here we show that soft substrates with elasticities commensurable to the elasticity of the brain promote the maturation of neural stem cell-derived neurons. In the absence of added growth factors, neurons differentiated on soft substrates displayed long neurites and presynaptic terminals, contrasting with the bipolar immature morphology of neurons differentiated on stiff substrates. Further, soft substrates supported an increase in astrocytic differentiation. However, stiffness cues could not override the dependency of astrocytic differentiation on Notch signaling. These results demonstrate that substrate elasticity per se can drive neuronal maturation thus defining a crucial parameter in neuronal differentiation of stem cells.


Subject(s)
Cell Differentiation/drug effects , Dimethylpolysiloxanes/pharmacology , Neurons/cytology , Neurons/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Shape/drug effects , Cells, Cultured , Neurites/drug effects , Neurites/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/drug effects , Synaptotagmins/metabolism
12.
Biomaterials ; 28(27): 3936-43, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17576007

ABSTRACT

Inkjet printing allows for the rapid and inexpensive printing of cells, materials, and protein molecules. However, the combination of inkjet printing and control of neural stem cell (NSC) multipotency and differentiation has remained unexplored. We used an inkjet printer (Canon BJC-2100) to print biologically active macromolecules on poly-acrylamide-based hydrogels (HydroGel(TM)), which were subsequently seeded with primary fetal NSCs. NSCs cultured on areas printed with fibroblast growth factor-2 (FGF2) remained undifferentiated, consistent with the effects of FGF2 when administered in solution. NSCs cultured in parallel on the same hydrogels but in areas printed with ciliary neurotrophic factor (CNTF) or fetal bovine serum (FBS) displayed a rapid induction of markers for astrocytic (glial fibrillary acidic protein, GFAP) or smooth muscle (smooth muscle actin, SMA) differentiation, respectively. These results are consistent with known actions of CNTF and FBS on NSCs. Importantly, NSCs cultured on a printed gradient of increasing levels of CNTF showed a linear increase in numbers of cells expressing GFAP, demonstrating a functional gradient of CNTF. Lastly, genetically modified NSCs proved to respond properly to printed macromolecules, suggesting that inkjet printing can successfully be combined with gene delivery to achieve effective control of stem cell differentiation.


Subject(s)
Intercellular Signaling Peptides and Proteins/administration & dosage , Neurons/cytology , Neurons/physiology , Printing/methods , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/methods , Animals , Animals, Newborn , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured , Computer Peripherals , Intercellular Signaling Peptides and Proteins/chemistry , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Tissue Engineering/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL