Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Genes Dev ; 28(21): 2407-20, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25367036

ABSTRACT

Cerebellar development is regulated by a coordinated spatiotemporal interplay between granule neuron progenitors (GNPs), Purkinje neurons, and glia. Abnormal development can trigger motor deficits, and more recent data indicate important roles in aspects of memory, behavior, and autism spectrum disorders (ASDs). Germline mutation in the NF1 tumor suppressor gene underlies Neurofibromatosis type 1, a complex disease that enhances susceptibility to certain cancers and neurological disorders, including intellectual deficits and ASD. The NF1 gene encodes for neurofibromin, a RAS GTPase-activating protein, and thus negatively regulates the RAS signaling pathway. Here, using mouse models to direct conditional NF1 ablation in either embryonic cerebellar progenitors or neonatal GNPs, we show that neurofibromin is required for appropriate development of cerebellar folia layering and structure. Remarkably, neonatal administration of inhibitors of the ERK pathway reversed the morphological defects. Thus, our findings establish a critical cell-autonomous role for the NF1-RAS-ERK pathway in the appropriate regulation of cerebellar development and provide a basis for using neonatal ERK inhibitor-based therapies to treat NF1-induced cerebellar disorders.


Subject(s)
Cerebellum/embryology , MAP Kinase Signaling System/physiology , Neurofibromin 1/metabolism , Stem Cells/cytology , Stem Cells/physiology , ras Proteins/physiology , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Cerebellum/cytology , Cerebellum/growth & development , Gene Deletion , Mice , Neurofibromin 1/genetics , Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Stem Cells/drug effects
2.
Ups J Med Sci ; 117(2): 99-112, 2012 May.
Article in English | MEDLINE | ID: mdl-22509804

ABSTRACT

The family of platelet-derived growth factors (PDGFs) plays a number of critical roles in normal embryonic development, cellular differentiation, and response to tissue damage. Not surprisingly, as it is a multi-faceted regulatory system, numerous pathological conditions are associated with aberrant activity of the PDGFs and their receptors. As we and others have shown, human gliomas, especially glioblastoma, express all PDGF ligands and both the two cell surface receptors, PDGFR-α and -ß. The cellular distribution of these proteins in tumors indicates that glial tumor cells are stimulated via PDGF/PDGFR-α autocrine and paracrine loops, while tumor vessels are stimulated via the PDGFR-ß. Here we summarize the initial discoveries on the role of PDGF and PDGF receptors in gliomas and provide a brief overview of what is known in this field.


Subject(s)
Brain Neoplasms/physiopathology , Glioma/physiopathology , Platelet-Derived Growth Factor/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Central Nervous System/physiology , Humans
3.
PLoS One ; 6(4): e18303, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21490965

ABSTRACT

BACKGROUND: Deregulation of platelet-derived growth factor (PDGF) signaling is a hallmark of malignant glioma. Two alternatively spliced PDGF-A mRNAs have been described, corresponding to a long (L) and a short (S) isoform of PDGF-A. In contrast to PDGF-A(S), the PDGF-A(L) isoform has a lysine and arginine rich carboxy-terminal extension that acts as an extracellular matrix retention motif. However, the exact role of PDGF-A(L) and how it functionally differs from the shorter isoform is not well understood. METHODOLOGY/PRINCIPAL FINDINGS: We overexpressed PDGF-A(L) as a transgene under control of the glial fibrillary acidic protein (GFAP) promoter in the mouse brain. This directs expression of the transgene to astrocytic cells and GFAP expressing neural stem cells throughout the developing and adult central nervous system. Transgenic mice exhibited a phenotype with enlarged skull at approximately 6-16 weeks of age and they died between 1.5 months and 2 years of age. We detected an increased number of undifferentiated cells in all areas of transgene expression, such as in the subependymal zone around the lateral ventricle and in the cerebellar medulla. The cells stained positive for Pdgfr-α, Olig2 and NG2 but this population did only partially overlap with cells positive for Gfap and the transgene reporter. Interestingly, a few mice presented with overt neoplastic glioma-like lesions composed of both Olig2 and Gfap positive cell populations and with microvascular proliferation, in a wild-type p53 background. CONCLUSIONS: Our findings show that PDGF-A(L) can induce accumulation of immature cells in the mouse brain. The strong expression of NG2, Pdgfr-α and Olig2 in PDGF-A(L) brains suggests that a fraction of these cells are oligodendrocyte progenitors. In addition, accumulation of fluid in the subarachnoid space and skull enlargement indicate that an increased intracranial pressure contributed to the observed lethality.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Glioma/metabolism , Glioma/pathology , Platelet-Derived Growth Factor/metabolism , Protein Isoforms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Glial Fibrillary Acidic Protein , Humans , Ki-67 Antigen/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Platelet-Derived Growth Factor/genetics , Protein Isoforms/genetics , SOXB1 Transcription Factors/metabolism
4.
J Oncol ; 2011: 852970, 2011.
Article in English | MEDLINE | ID: mdl-21209724

ABSTRACT

Malignant glioma is the most common brain tumor in adults and is associated with a very poor prognosis. Mutations in the p53 tumor suppressor gene are frequently detected in gliomas. p53 is well-known for its ability to induce cell cycle arrest, apoptosis, senescence, or differentiation following cellular stress. That the guardian of the genome also controls stem cell self-renewal and suppresses pluripotency adds a novel level of complexity to p53. Exactly how p53 works in order to prevent malignant transformation of cells in the central nervous system remains unclear, and despite being one of the most studied proteins, there is a need to acquire further knowledge about p53 in neural stem cells. Importantly, the characterization of glioma cells with stem-like properties, also known as brain tumor stem cells, has opened up for the development of novel targeted therapies. Here, we give an overview of what is currently known about p53 in brain tumors and neural stem cells. Specifically, we review the literature regarding transformation of adult neural stem cells and, we discuss how the loss of p53 and deregulation of growth factor signaling pathways, such as increased PDGF signaling, lead to brain tumor development. Reactivation of p53 in brain tumor stem cell populations in combination with current treatments for glioma should be further explored and may become a viable future therapeutic approach.

5.
Glia ; 57(11): 1143-53, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19115382

ABSTRACT

Glioblastomas are the most common and malignant astrocytic brain tumors in human adults. The tumor suppressor gene TP53 is commonly mutated and/or lost in astrocytic brain tumors and the TP53 alterations are often found in combination with excessive growth factor signaling via PDGF/PDGFRalpha. Here, we have generated transgenic mice over-expressing human PDGFB in brain, under control of the human GFAP promoter. These mice showed no phenotype, but on a Trp53 null background a majority of them developed brain tumors. This occurred at 2-6 months of age and tumors displayed human glioblastoma-like features with integrated development of Pdgfralpha+ tumor cells and Pdgfrbeta+/Nestin+ vasculature. The transgene was expressed in subependymal astrocytic cells, in glia limitans, and in astrocytes throughout the brain substance, and subsequently, microscopic tumor lesions were initiated equally in all these areas. With tumor size, there was an increase in Nestin positivity and variability in lineage markers. These results indicate an unexpected plasticity of all astrocytic cells in the adult brain, not only of SVZ cells. The results also indicate a contribution of widely distributed Pdgfralpha+ precursor cells in the tumorigenic process.


Subject(s)
Brain Neoplasms/genetics , Brain/metabolism , Genes, p53 , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-sis/metabolism , Animals , Astrocytes/metabolism , Brain/blood supply , Brain Neoplasms/metabolism , Disease Models, Animal , Gene Expression , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/metabolism , Humans , Intermediate Filament Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/metabolism , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...