Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Proc Natl Acad Sci U S A ; 120(16): e2222084120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37040416

ABSTRACT

Macrophage targeting therapies have had limited clinical success in glioblastoma (GBM). Further understanding the GBM immune microenvironment is critical for refining immunotherapeutic approaches. Here, we use genetically engineered mouse models and orthotopic transplantation-based GBM models with identical driver mutations and unique cells of origin to examine the role of tumor cell lineage in shaping the immune microenvironment and response to tumor-associated macrophage (TAM) depletion therapy. We show that oligodendrocyte progenitor cell lineage-associated GBMs (Type 2) recruit more immune infiltrates and specifically monocyte-derived macrophages than subventricular zone neural stem cell-associated GBMs (Type 1). We then devise a TAM depletion system that offers a uniquely robust and sustained TAM depletion. We find that extensive TAM depletion in these cell lineage-based GBM models affords no survival benefit. Despite the lack of survival benefit of TAM depletion, we show that Type 1 and Type 2 GBMs have unique molecular responses to TAM depletion. In sum, we demonstrate that GBM cell lineage influences TAM ontogeny and abundance and molecular response to TAM depletion.


Subject(s)
Brain Neoplasms , Glioblastoma , Mice , Animals , Tumor-Associated Macrophages/metabolism , Cell Lineage , Glioblastoma/pathology , Brain Neoplasms/pathology , Macrophages/metabolism , Neoplastic Processes , Tumor Microenvironment
2.
Cell Chem Biol ; 30(2): 214-229.e18, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36758549

ABSTRACT

Glioblastoma (GBM) is an aggressive adult brain cancer with few treatment options due in part to the challenges of identifying brain-penetrant drugs. Here, we investigated the mechanism of MM0299, a tetracyclic dicarboximide with anti-glioblastoma activity. MM0299 inhibits lanosterol synthase (LSS) and diverts sterol flux away from cholesterol into a "shunt" pathway that culminates in 24(S),25-epoxycholesterol (EPC). EPC synthesis following MM0299 treatment is both necessary and sufficient to block the growth of mouse and human glioma stem-like cells by depleting cellular cholesterol. MM0299 exhibits superior selectivity for LSS over other sterol biosynthetic enzymes. Critical for its application in the brain, we report an MM0299 derivative that is orally bioavailable, brain-penetrant, and induces the production of EPC in orthotopic GBM tumors but not normal mouse brain. These studies have implications for the development of an LSS inhibitor to treat GBM or other neurologic indications.


Subject(s)
Glioblastoma , Glioma , Adult , Humans , Lanosterol/pharmacology , Lanosterol/metabolism , Brain/metabolism , Glioma/drug therapy , Glioma/metabolism , Cholesterol , Glioblastoma/drug therapy
3.
Mol Ther ; 31(3): 810-824, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36463402

ABSTRACT

Activation of neurotrophic factor signaling is a promising therapy for neurodegeneration. However, the transient nature of ligand-dependent activation limits its effectiveness. In this study, we solved this problem by inventing a system that forces membrane localization of the intracellular domain of tropomyosin receptor kinase B (iTrkB), which results in constitutive activation without ligands. Our system overcomes the small size limitation of the genome packaging in adeno-associated virus (AAV) and allows high expression of the transgene. Using AAV-mediated gene therapy in the eyes, we demonstrate that iTrkB expression enhances neuroprotection in mouse models of glaucoma and stimulates robust axon regeneration after optic nerve injury. In addition, iTrkB expression in the retina was also effective in an optic tract transection model, in which the injury site is near the superior colliculus. Regenerating axons successfully formed pathways to their brain targets, resulting in partial recovery of visual behavior. Our system may also be applicable to other trophic factor signaling pathways and lead to a significant advance in the field of gene therapy for neurotrauma and neurodegenerative disorders, including glaucoma.


Subject(s)
Glaucoma , Retinal Ganglion Cells , Mice , Animals , Retinal Ganglion Cells/metabolism , Axons/physiology , Nerve Regeneration/genetics , Retina , Glaucoma/genetics , Glaucoma/therapy , Glaucoma/metabolism , Disease Models, Animal
4.
Dev Cell ; 57(1): 32-46.e8, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35016005

ABSTRACT

We test the hypothesis that glioblastoma harbors quiescent cancer stem cells that evade anti-proliferative therapies. Functional characterization of spontaneous glioblastomas from genetically engineered mice reveals essential quiescent stem-like cells that can be directly isolated from tumors. A derived quiescent cancer-stem-cell-specific gene expression signature is enriched in pre-formed patient GBM xenograft single-cell clusters that lack proliferative gene expression. A refined human 118-gene signature is preserved in quiescent single-cell populations from primary and recurrent human glioblastomas. The F3 cell-surface receptor mRNA, expressed in the conserved signature, identifies quiescent tumor cells by antibody immunohistochemistry. F3-antibody-sorted glioblastoma cells exhibit stem cell gene expression, enhance self-renewal in culture, drive tumor initiation and serial transplantation, and reconstitute tumor heterogeneity. Upon chemotherapy, the spared cancer stem cell pool becomes activated and accelerates transition to proliferation. These results help explain conventional treatment failure and lay a conceptual framework for alternative therapies.


Subject(s)
Cell Survival/physiology , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Division/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/pathology , Heterografts , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Transcriptome/genetics
5.
Curr Opin Neurol ; 34(6): 868-874, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34581301

ABSTRACT

PURPOSE OF REVIEW: Cancer stem cells (CSCs) have been implicated in the hierarchical heterogeneity and treatment resistance of hematologic and solid tumor malignancies, including gliomas, for several decades now but their therapeutic targeting has not been fully realized. Recent studies have uncovered deeper layers of CSC complexity, related to developmental origins, plasticity, cellular states, and interface with the microenvironment. RECENT FINDINGS: Sequencing and in-vivo lineage-tracing studies in mouse and patient-derived models show evidence of stem and progenitor origin of glioma, at the same time that genomic studies show a relatedness of glioma CSCs with radial glia. The spate of single-cell sequencing analyses demonstrates the diversity of transcriptional cellular states, which are susceptible to transitions, indicating the plasticity of glioma CSCs. The evolution of glioma CSCs and their interactions with niche cells play important roles in CSC treatment resistance and immune evasion, with epigenetic modulation as one of the emerging mechanisms. SUMMARY: To harness the potential of CSCs for clinical application, there is urgent need to investigate their complex nature and myriad interactions, to better understand the contribution of these self-renewing, stem-like cancer cells in the pathogenesis and therapy resistance of malignant brain tumors.


Subject(s)
Brain Neoplasms , Glioma , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Glioma/genetics , Glioma/therapy , Humans , Mice , Neoplastic Stem Cells , Tumor Microenvironment
6.
Cell Stem Cell ; 28(8): 1397-1410.e4, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34010628

ABSTRACT

NF1-associated malignant peripheral nerve sheath tumors (MPNSTs) are the major cause of mortality in neurofibromatosis. MPNSTs arise from benign peripheral nerve plexiform neurofibromas that originate in the embryonic neural crest cell lineage. Using reporter transgenes that label early neural crest lineage cells in multiple NF1 MPNST mouse models, we discover and characterize a rare MPNST cell population with stem-cell-like properties, including quiescence, that is essential for tumor initiation and relapse. Following isolation of these cells, we derive a cancer-stem-cell-specific gene expression signature that includes consensus embryonic neural crest genes and identify Nestin as a marker for the MPNST cell of origin. Combined targeting of cancer stem cells along with antimitotic chemotherapy yields effective tumor inhibition and prolongs survival. Enrichment of the cancer stem cell signature in cognate human tumors supports the generality and relevance of cancer stem cells to MPNST therapy development.


Subject(s)
Neurofibromatosis 1 , Neurofibrosarcoma , Animals , Disease Models, Animal , Mice , Neoplasm Recurrence, Local , Neurofibromatosis 1/genetics
7.
CRISPR J ; 4(1): 147-154, 2021 02.
Article in English | MEDLINE | ID: mdl-33567216

ABSTRACT

Mice derived entirely from embryonic stem (ES) cells can be generated through tetraploid complementation. Although XY male ES cell lines are commonly used in this system, occasionally, monosomic XO female mice are produced through spontaneous Y chromosome loss. Here, we describe an efficient method to obtain monosomic XO ES cells by CRISPR-Cas9-mediated deletion of the Y chromosome, allowing generation of female clonal mice by tetraploid complementation. The monosomic XO female mice are viable and able to produce normal male and female offspring. Direct generation of clonal mice in both sexes can significantly accelerate the production of complex genetically modified mouse models.


Subject(s)
CRISPR-Cas Systems , Chromosome Deletion , Embryonic Stem Cells , Infertility, Male , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development , Y Chromosome , Animals , Cell Line , Chromosomes, Human, Y , Clustered Regularly Interspaced Short Palindromic Repeats , Embryo, Mammalian , Female , Genotyping Techniques , Karyotyping , Male , Mice
8.
Clin Cancer Res ; 27(7): 1958-1966, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33495310

ABSTRACT

PURPOSE: Glioblastoma (GBM) is the most common malignant brain tumor in adults. Various immunotherapeutic approaches to improve patient survival are being developed, but the molecular mechanisms of immunotherapy resistance are currently unknown. Here, we explored the ability of a humanized radiolabeled CD8-targeted minibody to noninvasively quantify tumor-infiltrating CD8-positive (CD8+) T cells using PET. EXPERIMENTAL DESIGN: We generated a peripheral blood mononuclear cell (PBMC) humanized immune system (HIS) mouse model and quantified the absolute number of CD8+ T cells by flow cytometry relative to the [64Cu]Cu-NOTA-anti-CD8 PET signal. To evaluate a patient-derived orthotopic GBM HIS model, we intracranially injected cells into NOG mice, humanized cohorts with multiple HLA-matched PBMC donors, and quantified CD8+ tumor-infiltrating lymphocytes by IHC. To determine whether [64Cu]Cu-NOTA-anti-CD8 images brain parenchymal T-cell infiltrate in GBM tumors, we performed PET and autoradiography and subsequently stained serial sections of brain tumor tissue by IHC for CD8+ T cells. RESULTS: Nontumor-bearing NOG mice injected with human PBMCs showed prominent [64Cu]Cu-NOTA-anti-CD8 uptake in the spleen and minimal radiotracer localization to the normal brain. NOG mice harboring intracranial human GBMs yielded high-resolution PET images of tumor-infiltrating CD8+ T cells. Radiotracer retention correlated with CD8+ T-cell numbers in spleen and tumor tissue. Our study demonstrates the ability of [64Cu]Cu-NOTA-anti-CD8 PET to quantify peripheral and tumor-infiltrating CD8+ T cells in brain tumors. CONCLUSIONS: Human CD8+ T cells infiltrate an orthotopic GBM in a donor-dependent manner. Furthermore, [64Cu]Cu-NOTA-anti-CD8 quantitatively images both peripheral and brain parenchymal human CD8+ T cells.


Subject(s)
Brain Neoplasms/diagnostic imaging , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/metabolism , Glioblastoma/diagnostic imaging , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Lymphocytes, Tumor-Infiltrating/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Animals , Brain Neoplasms/immunology , Copper Radioisotopes , Female , Glioblastoma/immunology , Humans , Isotope Labeling , Mice
9.
Proc Natl Acad Sci U S A ; 117(49): 31448-31458, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229571

ABSTRACT

Adult neural stem cells (NSC) serve as a reservoir for brain plasticity and origin for certain gliomas. Lineage tracing and genomic approaches have portrayed complex underlying heterogeneity within the major anatomical location for NSC, the subventricular zone (SVZ). To gain a comprehensive profile of NSC heterogeneity, we utilized a well-validated stem/progenitor-specific reporter transgene in concert with single-cell RNA sequencing to achieve unbiased analysis of SVZ cells from infancy to advanced age. The magnitude and high specificity of the resulting transcriptional datasets allow precise identification of the varied cell types embedded in the SVZ including specialized parenchymal cells (neurons, glia, microglia) and noncentral nervous system cells (endothelial, immune). Initial mining of the data delineates four quiescent NSC and three progenitor-cell subpopulations formed in a linear progression. Further evidence indicates that distinct stem and progenitor populations reside in different regions of the SVZ. As stem/progenitor populations progress from neonatal to advanced age, they acquire a deficiency in transition from quiescence to proliferation. Further data mining identifies stage-specific biological processes, transcription factor networks, and cell-surface markers for investigation of cellular identities, lineage relationships, and key regulatory pathways in adult NSC maintenance and neurogenesis.


Subject(s)
Aging/genetics , Cell Lineage , Lateral Ventricles/anatomy & histology , Lateral Ventricles/cytology , Stem Cell Niche/genetics , Transcriptome/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Lineage/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transgenes
10.
Cancer Cell ; 38(3): 366-379.e8, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32649888

ABSTRACT

Glioblastoma, the predominant adult malignant brain tumor, has been computationally classified into molecular subtypes whose functional relevance remains to be comprehensively established. Tumors from genetically engineered glioblastoma mouse models initiated by identical driver mutations in distinct cells of origin portray unique transcriptional profiles reflective of their respective lineage. Here, we identify corresponding transcriptional profiles in human glioblastoma and describe patient-derived xenografts with species-conserved subtype-discriminating functional properties. The oligodendrocyte lineage-associated glioblastoma subtype requires functional ERBB3 and harbors unique therapeutic sensitivities. These results highlight the importance of cell lineage in glioblastoma independent of driver mutations and provide a methodology for functional glioblastoma classification for future clinical investigations.


Subject(s)
Brain Neoplasms/genetics , Cell Lineage/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Dasatinib/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Mice, Knockout , Mice, Nude , Oligodendroglia/cytology , Oligodendroglia/metabolism , Xenograft Model Antitumor Assays/methods
11.
Nature ; 567(7748): 341-346, 2019 03.
Article in English | MEDLINE | ID: mdl-30842654

ABSTRACT

Cancer-specific inhibitors that reflect the unique metabolic needs of cancer cells are rare. Here we describe Gboxin, a small molecule that specifically inhibits the growth of primary mouse and human glioblastoma cells but not that of mouse embryonic fibroblasts or neonatal astrocytes. Gboxin rapidly and irreversibly compromises oxygen consumption in glioblastoma cells. Gboxin relies on its positive charge to associate with mitochondrial oxidative phosphorylation complexes in a manner that is dependent on the proton gradient of the inner mitochondrial membrane, and it inhibits the activity of F0F1 ATP synthase. Gboxin-resistant cells require a functional mitochondrial permeability transition pore that regulates pH and thus impedes the accumulation of Gboxin in the mitochondrial matrix. Administration of a metabolically stable Gboxin analogue inhibits glioblastoma allografts and patient-derived xenografts. Gboxin toxicity extends to established human cancer cell lines of diverse organ origin, and shows that the increased proton gradient and pH in cancer cell mitochondria is a mode of action that can be targeted in the development of antitumour reagents.


Subject(s)
Glioblastoma/drug therapy , Glioblastoma/metabolism , Oxidative Phosphorylation/drug effects , Allografts , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hydrogen-Ion Concentration , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Neoplasm Transplantation , Organ Specificity , Proton-Motive Force/drug effects , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/metabolism , Xenograft Model Antitumor Assays
12.
Am J Med Genet A ; 179(6): 1091-1097, 2019 06.
Article in English | MEDLINE | ID: mdl-30908877

ABSTRACT

The neurofibromatoses, which include neurofibromatosis type I (NF1), neurofibromatosis type II (NF2), and schwannomatosis, are a group of syndromes characterized by tumor growth in the nervous system. The RASopathies are a group of syndromes caused by germline mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. The RASopathies include NF1, Noonan syndrome, Noonan syndrome with multiple lentigines, Costello syndrome, cardio-facio-cutaneous syndrome, Legius syndrome, capillary malformation arterio-venous malformation syndrome, and SYNGAP1 autism. Due to their common underlying pathogenetic etiology, all these syndromes have significant phenotypic overlap of which one common feature include a predisposition to tumors, which may be benign or malignant. Together as a group, they represent one of the most common multiple congenital anomaly syndromes estimating to affect approximately one in 1000 individuals worldwide. The subcontinent of India represents one of the largest populations in the world, yet remains underserved from an aspect of clinical genetics services. In an effort to bridge this gap, the First International Conference on RASopathies and Neurofibromatoses in Asia: Identification and Advances of New Therapeutics was held in Kochi, Kerala, India. These proceedings chronicle this timely and topical international symposium directed at discussing the best practices and therapies for individuals with neurofibromatoses and RASopathies.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Mitogen-Activated Protein Kinases/genetics , Neurofibromatoses/etiology , ras Proteins/genetics , Biomarkers , Disease Management , Genetic Association Studies/methods , Humans , Mitogen-Activated Protein Kinases/metabolism , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Neurofibromatoses/diagnosis , Neurofibromatoses/therapy , Signal Transduction , Translational Research, Biomedical , ras Proteins/metabolism
13.
Nat Neurosci ; 22(4): 545-555, 2019 04.
Article in English | MEDLINE | ID: mdl-30778149

ABSTRACT

The contribution of lineage identity and differentiation state to malignant transformation is controversial. We have previously shown that adult neural stem and early progenitor cells give origin to glioblastoma. Here we systematically assessed the tumor-initiating potential of adult neural populations at various stages of lineage progression. Cell type-specific tamoxifen-inducible Cre recombinase transgenes were used to target glioblastoma-relevant tumor suppressors Nf1, Trp53 and Pten in late-stage neuronal progenitors, neuroblasts and differentiated neurons. Mutant mice showed cellular and molecular defects demonstrating the impact of tumor suppressor loss, with mutant neurons being the most resistant to early changes associated with tumor development. However, we observed no evidence of glioma formation. These studies show that increasing lineage restriction is accompanied by decreasing susceptibility to malignant transformation, indicating a glioblastoma cell-of-origin hierarchy in which stem cells sit at the apex and differentiated cell types are least susceptible to tumorigenesis.


Subject(s)
Brain Neoplasms/metabolism , Cell Lineage , Glioblastoma/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Proliferation , Female , Male , Mice, Transgenic , Neurofibromin 1/metabolism , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Nat Commun ; 8(1): 1668, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29162814

ABSTRACT

Adult neurogenesis persists in the rodent dentate gyrus and is stimulated by chronic treatment with conventional antidepressants through BDNF/TrkB signaling. Ketamine in low doses produces both rapid and sustained antidepressant effects in patients. Previous studies have shed light on post-transcriptional synaptic NMDAR mediated mechanisms underlying the acute effect, but how ketamine acts at the cellular level to sustain this anti-depressive function for prolonged periods remains unclear. Here we report that ketamine accelerates differentiation of doublecortin-positive adult hippocampal neural progenitors into functionally mature neurons. This process requires TrkB-dependent ERK pathway activation. Genetic ablation of TrkB in neural stem/progenitor cells, or pharmacologic disruption of ERK signaling, or inhibition of adult neurogenesis, each blocks the ketamine-induced behavioral responses. Conversely, enhanced ERK activity via Nf1 gene deletion extends the response and rescues both neurogenic and behavioral deficits in mice lacking TrkB. Thus, TrkB-dependent neuronal differentiation is involved in the sustained antidepressant effects of ketamine.


Subject(s)
Cell Differentiation/drug effects , Ketamine/pharmacology , Neural Stem Cells/metabolism , Receptor, trkB/metabolism , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Female , Hippocampus/cytology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Receptor, trkB/genetics
16.
Br J Cancer ; 115(12): 1445-1450, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27832665

ABSTRACT

The cellular origin of gliomas remains a topic of controversy in cancer research. Advances in neurobiology, molecular genetics, and functional genomics have ushered new insights through exploiting the development of more sophisticated tools to address this question. Diverse distinct cell populations in the adult brain have been reported to give rise to gliomas, although how these studies relate physiologically to mechanisms of spontaneous tumour formation via accumulation of tumour-initiating mutations within a single cell are less well developed. Recent studies in animal models indicate that the lineage of the tumour-initiating cell may contribute to the biological and genomic phenotype of glioblastoma. These results suggest that the cell of origin may not only serve as a source of diversity for these tumours, but may also provide new avenues for improved diagnostics and therapeutic targeting that may prolong the lives of patients.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Animals , Brain Neoplasms/genetics , Disease Models, Animal , Glioma/genetics , Humans , Mice , Mutation
17.
Article in English | MEDLINE | ID: mdl-27815542

ABSTRACT

The cellular origins and the mechanisms of progression, maintenance of tumorigenicity, and therapeutic resistance are central questions in the glioblastoma multiforme (GBM) field. Using tumor suppressor mouse models, our group recently reported two independent populations of adult GBM-initiating central nervous system progenitors. We found different functional and molecular subtypes depending on the tumor-initiating cell lineage, indicating that the cell of origin is a driver of GBM subtype diversity. Using an in vivo model, we also showed that GBM cancer stem cells (CSCs) or glioma stem cells (GSCs) contribute to resistance to chemotherapeutic agents and that genetic ablation of GSCs leads to a delay in tumor progression. These studies are consistent with the cell of origin and CSCs as critical regulators of the pathogenesis of GBM.


Subject(s)
Cell Proliferation/physiology , Central Nervous System/cytology , Disease Models, Animal , Glioblastoma/pathology , Neoplastic Stem Cells/cytology , Animals , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Humans , Mice , Neoplastic Stem Cells/drug effects
18.
Cancer Cell ; 28(4): 429-440, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26461091

ABSTRACT

A central question in glioblastoma multiforme (GBM) research is the identity of the tumor-initiating cell, and its contribution to the malignant phenotype and genomic state. We examine the potential of adult lineage-restricted progenitors to induce fully penetrant GBM using CNS progenitor-specific inducible Cre mice to mutate Nf1, Trp53, and Pten. We identify two phenotypically and molecularly distinct GBM subtypes governed by identical driver mutations. We demonstrate that the two subtypes arise from functionally independent pools of adult CNS progenitors. Despite histologic identity as GBM, these tumor types are separable based on the lineage of the tumor-initiating cell. These studies point to the cell of origin as a major determinant of GBM subtype diversity.


Subject(s)
Adult Stem Cells/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Central Nervous System/cytology , Glioblastoma/genetics , Glioblastoma/pathology , Adult Stem Cells/metabolism , Animals , Cell Movement , Cell Proliferation , Humans , Mice , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurofibromin 1/genetics , PTEN Phosphohydrolase/genetics , Tumor Suppressor Protein p53/genetics
19.
J Immunol ; 195(1): 31-5, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002977

ABSTRACT

Ras GTPase-activating proteins (RasGAPs) inhibit signal transduction initiated through the Ras small GTP-binding protein. However, which members of the RasGAP family act as negative regulators of T cell responses is not completely understood. In this study, we investigated potential roles for the RasGAPs RASA1 and neurofibromin 1 (NF1) in T cells through the generation and analysis of T cell-specific RASA1 and NF1 double-deficient mice. In contrast to mice lacking either RasGAP alone in T cells, double-deficient mice developed T cell acute lymphoblastic leukemia/lymphoma, which originated at an early point in T cell development and was dependent on activating mutations in the Notch1 gene. These findings highlight RASA1 and NF1 as cotumor suppressors in the T cell lineage.


Subject(s)
Neurofibromin 1/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , p120 GTPase Activating Protein/genetics , Animals , Gene Deletion , Gene Expression Regulation , Mice , Mice, Knockout , Mutation , Neurofibromin 1/deficiency , Neurofibromin 1/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/immunology , Signal Transduction , Spleen/immunology , Spleen/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Time Factors , p120 GTPase Activating Protein/deficiency , p120 GTPase Activating Protein/immunology
20.
Article in English | MEDLINE | ID: mdl-25635044

ABSTRACT

Malignant glioma remains incurable despite tremendous advancement in basic research and clinical practice. The identification of the cell(s) of origin should provide deep insights into leverage points for one to halt disease progression. Here we summarize recent studies that support the notion that neural stem cell (NSC), astrocyte, and oligodendrocyte precursor cell (OPC) can all serve as the cell of origin. We also lay out important considerations on technical rigor for further exploring this subject. Finally, we share perspectives on how one could apply the knowledge of cell of origin to develop effective treatment methods. Although it will be a difficult battle, victory should be within reach as along as we continue to assimilate new information and facilitate the collaboration among basic scientists, translational researchers, and clinicians.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioma/pathology , Glioma/therapy , Astrocytes/pathology , Humans , Neural Stem Cells/pathology , Oligodendroglia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...