Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neuro Oncol ; 22(8): 1150-1161, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32296841

ABSTRACT

BACKGROUND: Imagining ways to prevent or treat glioblastoma (GBM) has been hindered by a lack of understanding of its pathogenesis. Although overexpression of platelet derived growth factor with two A-chains (PDGF-AA) may be an early event, critical details of the core biology of GBM are lacking. For example, existing PDGF-driven models replicate its microscopic appearance, but not its genomic architecture. Here we report a model that overcomes this barrier to authenticity. METHODS: Using a method developed to establish neural stem cell cultures, we investigated the effects of PDGF-AA on subventricular zone (SVZ) cells, one of the putative cells of origin of GBM. We microdissected SVZ tissue from p53-null and wild-type adult mice, cultured cells in media supplemented with PDGF-AA, and assessed cell viability, proliferation, genome stability, and tumorigenicity. RESULTS: Counterintuitive to its canonical role as a growth factor, we observed abrupt and massive cell death in PDGF-AA: wild-type cells did not survive, whereas a small fraction of null cells evaded apoptosis. Surviving null cells displayed attenuated proliferation accompanied by whole chromosome gains and losses. After approximately 100 days in PDGF-AA, cells suddenly proliferated rapidly, acquired growth factor independence, and became tumorigenic in immune-competent mice. Transformed cells had an oligodendrocyte precursor-like lineage marker profile, were resistant to platelet derived growth factor receptor alpha inhibition, and harbored highly abnormal karyotypes similar to human GBM. CONCLUSION: This model associates genome instability in neural progenitor cells with chronic exposure to PDGF-AA and is the first to approximate the genomic landscape of human GBM and the first in which the earliest phases of the disease can be studied directly.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Stem Cells , Platelet-Derived Growth Factor , Tumor Suppressor Protein p53 , Animals , Brain Neoplasms/chemically induced , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cells, Cultured , Glioblastoma/chemically induced , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Platelet-Derived Growth Factor/pharmacology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
3.
Glia ; 65(12): 1914-1926, 2017 12.
Article in English | MEDLINE | ID: mdl-28836293

ABSTRACT

High-grade gliomas (HGG), including glioblastomas, are characterized by invasive growth, resistance to therapy, and high inter- and intra-tumoral heterogeneity. The key histological hallmarks of glioblastoma are pseudopalisading necrosis and microvascular proliferation, which allow pathologists to distinguish glioblastoma from lower-grade gliomas. In addition to being genetically and molecularly heterogeneous, HGG are also heterogeneous with respect to the composition of their microenvironment. The question of whether this microenvironmental heterogeneity is driven by the molecular identity of the tumor remains controversial. However, this question is of utmost importance since microenvironmental, non-neoplastic cells are key components of the most radiotherapy- and chemotherapy-resistant niches of the tumor. Our work demonstrates a versatile, reliable, and reproducible adult HGG mouse model with NF1-silencing as a driver mutation. This model shows significant differences in tumor microenvironment, expression of subtype-specific markers, and response to standard therapy when compared to our established PDGFB-overexpressing HGG mouse model. PDGFB-overexpressing and NF1-silenced murine tumors closely cluster with human proneural and mesenchymal subtypes, as well as PDGFRA-amplified and NF1-deleted/mutant human tumors, respectively, at both the RNA and protein expression levels. These models can be generated in fully immunocompetent mixed or C57BL/6 genetic background mice, and therefore can easily be incorporated into preclinical studies for cancer cell-specific or immune cell-targeting drug discovery studies.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Mutation/genetics , Proto-Oncogene Proteins c-sis/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Cerebral Ventricles/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Disease Models, Animal , Doublecortin Domain Proteins , Gene Expression Regulation, Neoplastic/drug effects , Glioma/diagnostic imaging , Glioma/genetics , Glioma/therapy , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Neuropeptides/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Temozolomide
4.
Curr Med Chem ; 15(19): 1863-9, 2008.
Article in English | MEDLINE | ID: mdl-18691044

ABSTRACT

Prostaglandins (PGs) are potent autocrine and paracrine oxygenated lipid molecules that contribute appreciably to physiologic and pathophysiologic responses in almost all organs, including brain. Emerging data indicate that the PGs, and more specifically PGE2, play a central role in brain diseases including ischemic injury and several neurodegenerative diseases. Given concerns over the potential toxicity from protracted use of cyclooxygenase inhibitors in the elderly, attention is now focused on blocking PGE2 signaling that is mediated by interactions with four distinct G protein-coupled receptors, EP1-4, which are differentially expressed on neuronal and glial cells throughout the central nervous system. EP1 activation has been shown to mediate Ca2+-dependent neurotoxicity in ischemic injury. EP2 activation has been shown to mediate microglial-induced paracrine neurotoxicity as well as suppress microglia internalization of aggregated neurotoxic peptides. Animal models support the potential efficacy of targeting specific EP receptor subtypes in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and ischemic stroke. However promising these preclinical studies are, they have yet to be followed by clinical trials targeting any EP receptor in neurologic diseases.


Subject(s)
Dinoprostone/metabolism , Nervous System Diseases/metabolism , Animals , Binding Sites , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Humans , Ligands , Nervous System Diseases/drug therapy , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
5.
FASEB J ; 19(13): 1923-5, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16129700

ABSTRACT

Biochemical characterization of the major detergent-insoluble proteins that comprise hallmark histopathologic lesions initiated the molecular era of Alzheimer's disease (AD) research. Here, we reinvestigated detergent-insoluble proteins in AD using modern proteomic techniques. Using liquid chromatography (LC)-mass spectrometry (MS)-MS-based proteomics, we robustly identified 125 proteins in the detergent-insoluble fraction of late-onset AD (LOAD) temporal cortex that included several proteins critical to Abeta production, components of synaptic scaffolding, and products of genes linked to an increased risk of LOAD; we verified 15 of 15 of these proteins by Western blot. Following multiple analyses, we estimated that these represent ~80% of detergent-insoluble proteins in LOAD detectable by our method. Abeta, tau, and 7 of 8 other newly identified detergent-insoluble proteins were disproportionately increased in temporal cortex from patients with LOAD and AD derived from mutations in PSEN1 and PSEN2; all of these except tau were elevated in individuals with prodromal dementia, while none except Abeta were elevated in aged APPswe mice. These results are consistent with the amyloid hypothesis of AD and extend it to include widespread protein insolubility, not exclusively Abeta insolubility, early in AD pathogenesis even before the onset of clinical dementia.


Subject(s)
Alzheimer Disease/pathology , Detergents/pharmacology , Proteomics/methods , tau Proteins/chemistry , Alzheimer Disease/metabolism , Amyloid/chemistry , Animals , Blotting, Western , Brain/pathology , Chromatography, Liquid , Dementia/pathology , Dendritic Cells/metabolism , Detergents/metabolism , Disease Progression , Genes, Dominant , Humans , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Mutation , Presenilin-1 , Presenilin-2 , Protein Binding , Protein Interaction Mapping
6.
Neurology ; 64(7): 1258-60, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15824357

ABSTRACT

Spinocerebellar ataxia 14 (SCA14) is associated with missense mutations in the protein kinase C gamma gene (PRKCG), rather than a nucleotide repeat expansion. In this large-scale study of PRKCG in patients with ataxia, two new missense mutations, an in-frame deletion, and a possible splice site mutation were found and can now be added to the four previously described missense mutations. The genotype/phenotype correlations in these families are described.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation/genetics , Protein Kinase C/genetics , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Deletion , Genetic Testing , Genotype , Humans , Male , Middle Aged , Mutation, Missense/genetics , Phenotype , Protein Kinase C/chemistry , Protein Structure, Tertiary/genetics , RNA Splice Sites/genetics , Spinocerebellar Ataxias/physiopathology
7.
FASEB J ; 19(7): 869-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15746184

ABSTRACT

We performed proteomic analysis of neurofibrillary tangles (NFTs) obtained by laser capture microdissection from pyramidal neurons in hippocampal sector CA1 in patients with Alzheimer disease (AD) using liquid chromatography (LC)-mass spectrometry (MS)/MS. We discovered a total of 155 proteins in laser captured NFT's, 72 of which were identified by multiple unique peptides. Of these 72 proteins, 63 had previously unknown association with NFTs; one of these was glyceraldehyde-3-phosphate dehydrogenase (GAPDH). We validated by immunohistochemistry that GAPDH co-localized with the majority of NFTs as well as plaque-like structures in AD brain and was co-immunoprecipitated by antibodies to abnormal forms of tau in AD, but not tau from AD temporal cortex. Characterization of GAPDH showed that it, along with phosphorylated tau and Abeta peptides, was present in detergent-insoluble fractions from AD temporal cortex but not from age-matched controls. These data are the first proteomic investigation of NFTs. Moreover, our results validate this approach by demonstrating that GAPDH, a glycolytic and microtubule binding protein, not only co-localized to NFTs and immunoprecipitated with PHF-tau, but also is one of the few proteins known to undergo conversion to a detergent-insoluble form in AD.


Subject(s)
Alzheimer Disease/enzymology , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/analysis , Nerve Tissue Proteins/metabolism , Neurofibrillary Tangles/enzymology , Proteomics , Blotting, Western , Chromatography, Liquid , Detergents , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/chemistry , Hippocampus/chemistry , Humans , Immunohistochemistry , Mass Spectrometry , Protein Structure, Secondary , Solubility , Temporal Lobe/chemistry , Trypsin/metabolism , tau Proteins
8.
Am J Hum Genet ; 72(4): 839-49, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12644968

ABSTRACT

We report a nonepisodic autosomal dominant (AD) spinocerebellar ataxia (SCA) not caused by a nucleotide repeat expansion that is, to our knowledge, the first such SCA. The AD SCAs currently comprise a group of > or =16 genetically distinct neurodegenerative conditions, all characterized by progressive incoordination of gait and limbs and by speech and eye-movement disturbances. Six of the nine SCAs for which the genes are known result from CAG expansions that encode polyglutamine tracts. Noncoding CAG, CTG, and ATTCT expansions are responsible for three other SCAs. Approximately 30% of families with SCA do not have linkage to the known loci. We recently mapped the locus for an AD SCA in a family (AT08) to chromosome 19q13.4-qter. A particularly compelling candidate gene, PRKCG, encodes protein kinase C gamma (PKC gamma), a member of a family of serine/threonine kinases. The entire coding region of PRKCG was sequenced in an affected member of family AT08 and in a group of 39 unrelated patients with ataxia not attributable to trinucleotide expansions. Three different nonconservative missense mutations in highly conserved residues in C1, the cysteine-rich region of the protein, were found in family AT08, another familial case, and a sporadic case. The mutations cosegregated with disease in both families. Structural modeling predicts that two of these amino acid substitutions would severely abrogate the zinc-binding or phorbol ester-binding capabilities of the protein. Immunohistochemical studies on cerebellar tissue from an affected member of family AT08 demonstrated reduced staining for both PKC gamma and ataxin 1 in Purkinje cells, whereas staining for calbindin was preserved. These results strongly support a new mechanism for neuronal cell dysfunction and death in hereditary ataxias and suggest that there may be a common pathway for PKC gamma-related and polyglutamine-related neurodegeneration.


Subject(s)
Mutation, Missense , Polymorphism, Genetic , Protein Kinase C/genetics , Spinocerebellar Ataxias/genetics , Amino Acid Sequence , Conserved Sequence , Female , Genes, Dominant , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Male , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Conformation , Protein Kinase C/chemistry , Reference Values , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...