Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Sci Rep ; 11(1): 24345, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934133

ABSTRACT

Mutations in the NF1 tumor suppressor gene are linked to arteriopathy. Nf1 heterozygosity (Nf1+/-) results in robust neointima formation, similar to humans, and myeloid-restricted Nf1+/- recapitulates this phenotype via MEK-ERK activation. Here we define the contribution of myeloid subpopulations to NF1 arteriopathy. Neutrophils from WT and Nf1+/- mice were functionally assessed in the presence of MEK and farnesylation inhibitors in vitro and neutrophil recruitment to lipopolysaccharide was assessed in WT and Nf1+/- mice. Littermate 12-15 week-old male wildtype and Nf1+/- mice were subjected to carotid artery ligation and provided either a neutrophil depleting antibody (1A8), liposomal clodronate to deplete monocytes/macrophages, or PD0325901 and neointima size was assessed 28 days after injury. Bone marrow transplant experiments assessed monocyte/macrophage mobilization during neointima formation. Nf1+/- neutrophils exhibit enhanced proliferation, migration, and adhesion via p21Ras activation of MEK in vitro and in vivo. Neutrophil depletion suppresses circulating Ly6Clow monocytes and enhances neointima size, while monocyte/macrophage depletion and deletion of CCR2 in bone marrow cells abolish neointima formation in Nf1+/- mice. Taken together, these findings suggest that neurofibromin-MEK-ERK activation in circulating neutrophils and monocytes during arterial remodeling is nuanced and points to important cross-talk between these populations in the pathogenesis of NF1 arteriopathy.


Subject(s)
Carotid Artery Injuries/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Myeloid Progenitor Cells/pathology , Neointima/pathology , Neurofibromatosis 1/pathology , Neurofibromin 1/physiology , Receptors, CCR2/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myeloid Progenitor Cells/metabolism , Neointima/etiology , Neointima/metabolism , Neurofibromatosis 1/etiology , Neurofibromatosis 1/metabolism
2.
Proc Natl Acad Sci U S A ; 116(44): 22122-22131, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611389

ABSTRACT

KRAS mutations occur in ∼35% of colorectal cancers and promote tumor growth by constitutively activating the mitogen-activated protein kinase (MAPK) pathway. KRAS mutations at codons 12, 13, or 61 are thought to prevent GAP protein-stimulated GTP hydrolysis and render KRAS-mutated colorectal cancers unresponsive to epidermal growth factor receptor (EGFR) inhibitors. We report here that KRAS G13-mutated cancer cells are frequently comutated with NF1 GAP but NF1 is rarely mutated in cancers with KRAS codon 12 or 61 mutations. Neurofibromin protein (encoded by the NF1 gene) hydrolyzes GTP directly in complex with KRAS G13D, and KRAS G13D-mutated cells can respond to EGFR inhibitors in a neurofibromin-dependent manner. Structures of the wild type and G13D mutant of KRAS in complex with neurofibromin (RasGAP domain) provide the structural basis for neurofibromin-mediated GTP hydrolysis. These results reveal that KRAS G13D is responsive to neurofibromin-stimulated hydrolysis and suggest that a subset of KRAS G13-mutated colorectal cancers that are neurofibromin-competent may respond to EGFR therapies.


Subject(s)
Colorectal Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , Guanosine Triphosphate/metabolism , Neurofibromin 1/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Catalytic Domain , Cell Line , Colorectal Neoplasms/drug therapy , GTPase-Activating Proteins/metabolism , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Models, Molecular , Neurofibromin 1/metabolism , Neurofibromin 1/physiology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics
3.
Article in English | MEDLINE | ID: mdl-30104198

ABSTRACT

Ras-specific GTPase-activating proteins (RasGAPs) down-regulate the biological activity of Ras proteins by accelerating their intrinsic rate of GTP hydrolysis, basically by a transition state stabilizing mechanism. Oncogenic Ras is commonly not sensitive to RasGAPs caused by interference of mutants with the electronic or steric requirements of the transition state, resulting in up-regulation of activated Ras in respective cells. RasGAPs are modular proteins containing a helical catalytic RasGAP module surrounded by smaller domains that are frequently involved in the subcellular localization or contributing to regulatory features of their host proteins. In this review, we summarize current knowledge about RasGAP structure, mechanism, regulation, and dual-substrate specificity and discuss in some detail neurofibromin, one of the most important negative Ras regulators in cellular growth control and neuronal function.


Subject(s)
ras GTPase-Activating Proteins/chemistry , Cell Enlargement , Down-Regulation/physiology , Enzyme Activation/physiology , GTP Phosphohydrolases/metabolism , Gap Junctions/physiology , Humans , Molecular Structure , Neurofibromin 1/physiology , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/physiology
4.
Sci Rep ; 8(1): 15381, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337665

ABSTRACT

Increasing evidence supports a close relationship between Ras-ERK1/2 activation in the striatum and L-DOPA-induced dyskinesia (LID). ERK1/2 activation by L-DOPA takes place through the crosstalk between D1R/AC/PKA/DARPP-32 pathway and NMDA/Ras pathway. Compelling genetic and pharmacological evidence indicates that Ras-ERK1/2 inhibition prevents LID onset and may even revert already established dyskinetic symptoms. However, it is currently unclear whether exacerbation of Ras-ERK1/2 activity in the striatum may further aggravate dyskinesia in experimental animal models. Here we took advantage of two genetic models in which Ras-ERK1/2 signaling is hyperactivated, the Nf1+/- mice, in which the Ras inhibitor neurofibromin is reduced, and the Ras-GRF1 overexpressing (Ras-GRF1 OE) transgenic mice in which a specific neuronal activator of Ras is enhanced. Nf1+/- and Ras-GRF1 OE mice were unilaterally lesioned with 6-OHDA and treated with an escalating L-DOPA dosing regimen. In addition, a subset of Nf1+/- hemi-parkinsonian animals was also co-treated with the Ras inhibitor lovastatin. Our results revealed that Nf1+/- and Ras-GRF1 OE mice displayed similar dyskinetic symptoms to their wild-type counterparts. This observation was confirmed by the lack of differences between mutant and wild-type mice in striatal molecular changes associated to LID (i.e., FosB, and pERK1/2 expression). Interestingly, attenuation of Ras activity with lovastatin does not weaken dyskinetic symptoms in Nf1+/- mice. Altogether, these data suggest that ERK1/2-signaling activation in dyskinetic animals is maximal and does not require further genetic enhancement in the upstream Ras pathway. However, our data also demonstrate that such a genetic enhancement may reduce the efficacy of anti-dyskinetic drugs like lovastatin.


Subject(s)
Dyskinesia, Drug-Induced/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Levodopa/toxicity , Lovastatin/pharmacology , Neurofibromin 1/physiology , ras Proteins/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dopamine Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Signal Transduction , ras Proteins/genetics
5.
JCI Insight ; 3(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29925695

ABSTRACT

Loss of the NF1 tumor suppressor gene causes the autosomal dominant condition, neurofibromatosis type 1 (NF1). Children and adults with NF1 suffer from pathologies including benign and malignant tumors to cognitive deficits, seizures, growth abnormalities, and peripheral neuropathies. NF1 encodes neurofibromin, a Ras-GTPase activating protein, and NF1 mutations result in hyperactivated Ras signaling in patients. Existing NF1 mutant mice mimic individual aspects of NF1, but none comprehensively models the disease. We describe a potentially novel Yucatan miniswine model bearing a heterozygotic mutation in NF1 (exon 42 deletion) orthologous to a mutation found in NF1 patients. NF1+/ex42del miniswine phenocopy the wide range of manifestations seen in NF1 patients, including café au lait spots, neurofibromas, axillary freckling, and neurological defects in learning and memory. Molecular analyses verified reduced neurofibromin expression in swine NF1+/ex42del fibroblasts, as well as hyperactivation of Ras, as measured by increased expression of its downstream effectors, phosphorylated ERK1/2, SIAH, and the checkpoint regulators p53 and p21. Consistent with altered pain signaling in NF1, dysregulation of calcium and sodium channels was observed in dorsal root ganglia expressing mutant NF1. Thus, these NF1+/ex42del miniswine recapitulate the disease and provide a unique, much-needed tool to advance the study and treatment of NF1.


Subject(s)
Disease Models, Animal , Neurofibromatosis 1 , Neurofibromin 1/metabolism , Swine , Animals , Cafe-au-Lait Spots , Exons/genetics , Fibroblasts/metabolism , GTPase-Activating Proteins/genetics , Ganglia, Spinal/metabolism , Gene Deletion , Gene Expression Regulation , Gene Knockout Techniques , Humans , Ion Channels , Learning , Male , Memory , Mutation , Neurofibroma , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Neurofibromin 1/physiology , Signal Transduction
6.
Mol Genet Metab ; 123(4): 518-525, 2018 04.
Article in English | MEDLINE | ID: mdl-29477258

ABSTRACT

Neurofibromatosis Type 1 (NF1) is a common autosomal dominant genetic disorder While NF1 is primarily associated with predisposition for tumor formation, muscle weakness has emerged as having a significant impact on quality of life. NF1 inactivation is linked with a canonical upregulation Ras-MEK-ERK signaling. This in this study we tested the capacity of the small molecule MEK inhibitor PD0325901 to influence the intramyocellular lipid accumulation associated with NF1 deficiency. Established murine models of tissue specific Nf1 deletion in skeletal muscle (Nf1MyoD-/-) and limb mesenchyme (Nf1Prx1-/-) were tested. Developmental PD0325901 dosing of dams pregnant with Nf1MyoD-/- progeny rescued the phenotype of day 3 pups including body weight and lipid accumulation by Oil Red O staining. In contrast, PD0325901 treatment of 4 week old Nf1Prx1-/- mice for 8 weeks had no impact on body weight, muscle wet weight, activity, or intramyocellular lipid. Examination of day 3 Nf1Prx1-/- pups showed differences between the two tissue-specific knockout strains, with lipid staining greatest in Nf1MyoD-/- mice, and fibrosis higher in Nf1Prx1-/- mice. These data show that a MEK/ERK dependent mechanism underlies NF1 muscle metabolism during development. However, crosstalk from Nf1-deficient non-muscle mesenchymal cells may impact upon muscle metabolism and fibrosis in neonatal and mature myofibers.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Extremities/pathology , Muscle, Skeletal/pathology , Muscular Diseases/prevention & control , Neurofibromatosis 1/physiopathology , Neurofibromin 1/physiology , Animals , Animals, Newborn , Diphenylamine/pharmacology , Female , Homeodomain Proteins/physiology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Muscular Diseases/pathology , MyoD Protein/physiology , Signal Transduction , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
7.
Int J Dev Biol ; 61(8-9): 531-536, 2017.
Article in English | MEDLINE | ID: mdl-29139538

ABSTRACT

Neurofibromatosis Type 1 (NF1) is an autosomal dominant genetic disorder that results in a variety of characteristic manifestations. Prior studies have shown reduced muscle size and global skeletal muscle weakness in children with NF1. This associated weakness can lead to significant challenges impacting on quality of life. Pre-clinical studies using a muscle-specific NF1 knockout mouse have linked this weakness to an underlying primary metabolic deficiency in the muscle. However, the neonatal lethality of this strain prevents analysis of the role of NF1 in adult muscle. In this study, we present the characterization of an inducible muscle-specific NF1 knockout strain (Nf1Pax7i f/f ) produced by cross breeding the Pax7-CreERT2 strain with the conditional Nf1flox/flox line. Tamoxifen dosing of 8-week old Nf1Pax7i f/f mice led to recombination of the floxed allele in muscle, as detected by PCR. Detailed phenotypic analysis of treated adult mice over 8 weeks revealed no changes in bodyweight or muscle weight, no histological signs of myopathy, and no functional evidence of distress or impairment. Subsequent analysis using the Ai9 Cre-dependent tdTomato reporter strain was used to analyse labelling in embryos and in adult mice. Cell tracking studies identified a lower than expected rate of integration of recombined satellite cells into adult muscle. In contrast, a high persistent contribution of embryonic cells that were Pax7+ were found in adult muscle. These findings indicate important caveats with the use of the Pax7-CreER T2 strain and highlight a need to develop new tools for investigating the function of NF1 in mature muscle.


Subject(s)
Cell Lineage , Muscle Development/physiology , Muscular Diseases/etiology , Neurofibromin 1/physiology , PAX7 Transcription Factor/physiology , Transgenes/physiology , Animals , Female , Mice , Mice, Knockout , Mice, Transgenic , Muscle Development/drug effects , Muscular Diseases/metabolism , Muscular Diseases/pathology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology
8.
Neuro Oncol ; 19(6): 808-819, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28039362

ABSTRACT

Background: Optic gliomas arising in the neurofibromatosis type 1 (NF1) cancer predisposition syndrome cause reduced visual acuity in 30%-50% of affected children. Since human specimens are rare, genetically engineered mouse (GEM) models have been successfully employed for preclinical therapeutic discovery and validation. However, the sequence of cellular and molecular events that culminate in retinal dysfunction and vision loss has not been fully defined relevant to potential neuroprotective treatment strategies. Methods: Nf1flox/mut GFAP-Cre (FMC) mice and age-matched Nf1flox/flox (FF) controls were euthanized at defined intervals from 2 weeks to 24 weeks of age. Optic nerve volumes were measured, and optic nerves/retinae analyzed by immunohistochemistry. Optical coherence tomography (OCT) was performed on anesthetized mice. FMC mice were treated with lovastatin from 12 to 16 weeks of age. Results: The earliest event in tumorigenesis was a persistent elevation in proliferation (4 wk), which preceded sustained microglia numbers and incremental increases in S100+ glial cells. Microglia activation, as evidenced by increased interleukin (IL)-1ß expression and morphologic changes, coincided with axonal injury and retinal ganglion cell (RGC) apoptosis (6 wk). RGC loss and retinal nerve fiber layer (RNFL) thinning then ensued (9 wk), as revealed by direct measurements and live-animal OCT. Lovastatin administration at 12 weeks prevented further RGC loss and RNFL thinning both immediately and 8 weeks after treatment completion. Conclusion: By defining the chronology of the cellular and molecular events associated with optic glioma pathogenesis, we demonstrate critical periods for neuroprotective intervention and visual preservation, as well as establish OCT as an accurate biomarker of RGC loss.


Subject(s)
Lovastatin/pharmacology , Neurofibromatosis 1/pathology , Neurofibromin 1/physiology , Optic Nerve Glioma/pathology , Retinal Ganglion Cells/pathology , Animals , Anticholesteremic Agents/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/metabolism , Optic Nerve Glioma/drug therapy , Optic Nerve Glioma/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Tomography, Optical Coherence , Visual Acuity
9.
Oncotarget ; 7(7): 7403-14, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26859681

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive sarcomas that arise sporadically or in association with the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome. In individuals with NF1, MPNSTs are hypothesized to arise from Nf1-deficient Schwann cell precursor cells following the somatic acquisition of secondary cooperating genetic mutations (e.g., p53 loss). To model this sequential genetic cooperativity, we coupled somatic lentivirus-mediated p53 knockdown in the adult right sciatic nerve with embryonic Schwann cell precursor Nf1 gene inactivation in two different Nf1 conditional knockout mouse strains. Using this approach, ~60% of mice with Periostin-Cre-mediated Nf1 gene inactivation (Periostin-Cre; Nf1(flox/flox) mice) developed tumors classified as low-grade MPNSTs following p53 knockdown (mean, 6 months). Similarly, ~70% of Nf1+/- mice with GFAP-Cre-mediated Nf1 gene inactivation (GFAP-Cre; Nf1(flox/null) mice) developed low-grade MPNSTs following p53 knockdown (mean, 3 months). In addition, wild-type and Nf1+/- mice with GFAP-Cre-mediated Nf1 loss develop MPNSTs following somatic p53 knockout with different latencies, suggesting potential influences of Nf1+/- stromal cells in MPNST pathogenesis. Collectively, this new MPNST model system permits the analysis of somatically-acquired events as well as tumor microenvironment signals that potentially cooperate with Nf1 loss in the development and progression of this deadly malignancy.


Subject(s)
Embryonic Stem Cells/pathology , Neurilemmoma/pathology , Neurofibromin 1/physiology , Schwann Cells/pathology , Sciatic Nerve/pathology , Tumor Suppressor Protein p53/physiology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Embryonic Stem Cells/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurilemmoma/genetics , Schwann Cells/metabolism , Sciatic Nerve/metabolism
10.
Postepy Hig Med Dosw (Online) ; 69: 1331-48, 2015 Dec 09.
Article in Polish | MEDLINE | ID: mdl-26671924

ABSTRACT

Neurofibromatosis type I (NF1) is multisystemic disease characterized by pigmentary skin changes, increased susceptibility to tumor formation, neurological deficits and skeletal defects. The disease is a monogenic, autosomal dominant disorder, caused by the presence of mutations in the NF1 gene encoding neurofibromin - a multifunctional regulatory protein. The basic function of neurofibromin protein is modulation of the RAS protein activity necessary for regulation of cell proliferation and differentiation by the RAS/MAPK and RAS/PI3K/AKT signal transduction pathways. In addition, neurofibromin is a regulator of adenylate cyclase activity and therefore may interfere with signaling by the cAMP/protein kinase A pathway that regulates cell cycle progression or learning and memory formation processes. Neurofibromin also interacts with many other proteins that are engaged in intracellular transport (tubulin, kinesin), actin cytoskeleton rearrangements (LIMK2, Rho and Rac) or morphogenesis of neural cells (syndecans, CRMP proteins). The activity of neurofibromin is strictly regulated by the expression of different NF1 mRNA isoforms depending on tissue type or period in organism development, the protein localization, posttranslational modifications (phosphorylation, ubiquitination) or interactions with other proteins (e.g. 14-3-3). The fact that neurofibromin is engaged in many cellular processes has significant consequences when the proper protein functioning is impaired due to decreased protein level or activity. It affects the normal cell function and results in disturbances of organism development that lead to the occurrence of clinical signs specific for NF1. In the article, the basic neurofibromin functions are presented in the context of the molecular pathogenesis of NF1.


Subject(s)
Mutation , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Signal Transduction , Alternative Splicing , Humans , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Neurofibromin 1/physiology , Protein Conformation , Protein Processing, Post-Translational
12.
Adv Biol Regul ; 55: 1-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24814062

ABSTRACT

The RAS pathway is one of the most commonly deregulated pathways in human cancer. Mutations in RAS genes occur in nearly 30% of all human tumors. However in some tumor types RAS mutations are conspicuously absent or rare, despite the fact that RAS and downstream effector pathways are hyperactivated. Recently, RAS GTPase Activating Proteins (RAS GAPs) have emerged as an expanding class of tumor suppressors that, when inactivated, provide an alternative mechanism of activating RAS. RAS GAPs normally turn off RAS by catalyzing the hydrolysis of RAS-GTP. As such, the loss of a RAS GAP would be expected to promote excessive RAS activation. Indeed, this is the case for the NF1 gene, which plays an established role in a familial tumor predisposition syndrome and a variety of sporadic cancers. However, there are 13 additional RAS GAP family members in the human genome. We are only now beginning to understand why there are so many RAS GAPs, how they differentially function, and what their potential role(s) in human cancer are. This review will focus on our current understanding of RAS GAPs in human disease and will highlight important outstanding questions.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , ras GTPase-Activating Proteins/physiology , Carrier Proteins/physiology , Drug Resistance, Neoplasm , GTPase-Activating Proteins/physiology , Genes, Neurofibromatosis 1/physiology , Guanine Nucleotide Exchange Factors/physiology , Humans , Neurofibromin 1/physiology , Tumor Suppressor Proteins/physiology , ras GTPase-Activating Proteins/genetics
13.
Pathol Biol (Paris) ; 62(2): 118-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24650525

ABSTRACT

This is a short review of neurofibromatosis-1 or von Recklinghausen's disease, due to a loss of function mutation of the gene neurofibromin-1, which normally inhibits the Ras MAPK-pathways. Among its symptoms, the strong oversynthesis of several collagen types designates this disease as producing a deregulation of extracellular matrix biosynthesis involved in tumor formation. Up to about 40% of the skin tumors consist of collagens. A short summary of the clinical manifestations and pathological and genetic mechanisms are also described.


Subject(s)
Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix/metabolism , Neurofibromatosis 1/metabolism , Skin Neoplasms/metabolism , Collagen/biosynthesis , Collagen/genetics , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Genes, Neurofibromatosis 1 , Humans , Neoplasm Proteins/metabolism , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Neurofibromin 1/physiology , Phenotype , Signal Transduction/physiology , Skin Neoplasms/genetics , Stromal Cells/metabolism , Tumor Microenvironment , ras Proteins/physiology
14.
J Neurosci ; 34(3): 776-83, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431436

ABSTRACT

In dendritic spines, Ras plays a critical role in synaptic plasticity but its regulation mechanism is not fully understood. Here, using a fluorescence resonance energy transfer/fluorescence lifetime imaging microscopy-based Ras imaging technique in combination with 2-photon glutamate uncaging, we show that neurofibromin, in which loss-of-function mutations cause Neurofibromatosis Type 1 (NF1), contributes to the majority (∼90%) of Ras inactivation in dendritic spines of pyramidal neurons in the CA1 region of the rat hippocampus. Loss of neurofibromin causes sustained Ras activation in spines, which leads to impairment of spine structural plasticity and loss of spines in an activity-dependent manner. Therefore, deregulation of postsynaptic Ras signaling may explain, at least in part, learning disabilities associated with NF1.


Subject(s)
Dendritic Spines/physiology , Genes, ras/physiology , Neurofibromin 1/physiology , Animals , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/genetics , Female , Hippocampus/physiology , Hippocampus/ultrastructure , Long-Term Potentiation/physiology , Male , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
15.
PLoS One ; 8(6): e66024, 2013.
Article in English | MEDLINE | ID: mdl-23762458

ABSTRACT

Children with neurofibromatosis type 1 (NF1) frequently have cognitive and behavioral deficits. Some of these deficits have been successfully modeled in Nf1 genetically-engineered mice that develop optic gliomas (Nf1 OPG mice). In the current study, we show that abnormal motivational influences affect the behavior of Nf1 OPG mice, particularly with regard to their response to novel environmental stimuli. For example, Nf1 OPG mice made fewer spontaneous alternations in a Y-maze and fewer arm entries relative to WT controls. However, analysis of normalized alternation data demonstrated that these differences were not due to a spatial working memory deficit. Other reported behavioral results (e.g., open-field test, below) suggest that differential responses to novelty and/or other motivational influences may be more important determinants of these kinds of behavior than simple differences in locomotor activity/spontaneous movements. Importantly, normal long-term depression was observed in hippocampal slices from Nf1 OPG mice. Results from elevated plus maze testing showed that differences in exploratory activity between Nf1 OPG and WT control mice may be dependent on the environmental context (e.g., threatening or non-threatening) under which exploration is being measured. Nf1 OPG mice also exhibited decreased exploratory hole poking in a novel holeboard and showed abnormal olfactory preferences, although L-dopa (50 mg/kg) administration resolved the abnormal olfactory preference behaviors. Nf1 OPG mice displayed an attenuated response to a novel open field in terms of decreased ambulatory activity and rearing but only during the first 10 min of the session. Importantly, Nf1 OPG mice demonstrated investigative rearing deficits with regard to a novel hanging object suspended on one side of the field which were not rescued by L-dopa administration. Collectively, our results provide new data important for evaluating therapeutic treatments aimed at ameliorating NF1-associated cognitive/behavioral deficits.


Subject(s)
Behavior, Animal/drug effects , Disease Models, Animal , Dopamine Agents/administration & dosage , Levodopa/administration & dosage , Motivation/drug effects , Neurofibromatosis 1/drug therapy , Neurofibromin 1/physiology , Animals , Depression/diagnosis , Depression/drug therapy , Depression/etiology , Electrophysiology , Female , Humans , Integrases/metabolism , Male , Maze Learning/drug effects , Memory Disorders/diagnosis , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Knockout , Mice, Transgenic , Neurofibromatosis 1/complications , Neurofibromatosis 1/metabolism
16.
Mol Immunol ; 55(3-4): 292-302, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23522726

ABSTRACT

TCR-mediated activation of the Ras signaling pathway is critical for T cell development in the thymus and function in the periphery. However, which members of a family of Ras GTPase-activating proteins (RasGAPs) negatively regulate Ras activation in T cells is unknown. In this study we examined a potential function for the neurofibromin 1 (NF1) RasGAP in the T cell lineage with the use of T cell-specific NF1-deficient mice. Surprisingly, on an MHC class I-restricted TCR transgenic background, NF1 was found to promote thymocyte positive selection. By contrast, NF1 neither promoted nor inhibited the negative selection of thymocytes. In the periphery, NF1 was found to be necessary for the maintenance of normal numbers of naïve CD4⁺ and CD8⁺ T cells but was dispensable as a regulator of TCR-induced Ras activation, cytokine synthesis, proliferation and differentiation and death. These findings point to a novel unexpected role for NF1 in T cell development as well as a regulator of T cell homeostasis.


Subject(s)
Cell Differentiation/immunology , Neurofibromin 1/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , ras Proteins/metabolism , Animals , Clonal Selection, Antigen-Mediated/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology
17.
Sci Signal ; 6(264): re1, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23443682

ABSTRACT

Inactivation of the small guanosine triphosphate-binding protein Ras during receptor signal transduction is mediated by Ras guanosine triphosphatase (GTPase)-activating proteins (RasGAPs). Ten different RasGAPs have been identified and have overlapping patterns of tissue distribution. However, genetic analyses are revealing critical nonredundant functions for each RasGAP in tissue homeostasis and as regulators of disease processes in mouse and man. Here, we discuss advances in understanding the role of RasGAPs in the maintenance of tissue integrity.


Subject(s)
Homeostasis/physiology , Models, Biological , Signal Transduction/physiology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/physiology , ras Proteins/physiology , Animals , Cell Membrane/metabolism , Humans , Mice , Neurofibromin 1/physiology , Protein Structure, Tertiary , p120 GTPase Activating Protein/physiology
18.
PLoS One ; 7(11): e49222, 2012.
Article in English | MEDLINE | ID: mdl-23145129

ABSTRACT

Neurofibromatosis is a well known familial tumor syndrome, however these patients also suffer from a number of vascular anomalies. The loss of NFl from the endothelium is embryonically lethal in mouse developmental models, however little is known regarding the molecular regulation by NF1 in endothelium. We investigated the consequences of losing NF1 expression on the function of endothelial cells using shRNA. The loss of NF1 was sufficient to elevate levels of active Ras under non-stimulated conditions. These elevations in Ras activity were associated with activation of downstream signaling including activation of ERK, AKT and mTOR. Cells knocked down in NF1 expression exhibited no cellular senescence. Rather, they demonstrated augmented proliferation and autonomous entry into the cell cycle. These proliferative changes were accompanied by enhanced expression of cyclin D, phosphorylation of p27(KIP), and decreases in total p27(KIP) levels, even under growth factor free conditions. In addition, NF1-deficient cells failed to undergo normal branching morphogenesis in a co-culture assay, instead forming planar islands with few tubules and branches. We find the changes induced by the loss of NF1 could be mitigated by co-expression of the GAP-related domain of NF1 implicating Ras regulation in these effects. Using doxycycline-inducible shRNA, targeting NF1, we find that the morphogenic changes are reversible. Similarly, in fully differentiated and stable vascular-like structures, the silencing of NF1 results in the appearance of abnormal vascular structures. Finally, the proliferative changes and the abnormal vascular morphogenesis are normalized by low-dose rapamycin treatment. These data provide a detailed analysis of the molecular and functional consequences of NF1 loss in human endothelial cells. These insights may provide new approaches to therapeutically addressing vascular abnormalities in these patients while underscoring a critical role for normal Ras regulation in maintaining the health and function of the vasculature.


Subject(s)
Cell Proliferation , Neurofibromin 1/genetics , Cell Cycle/genetics , Cellular Senescence/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Knockdown Techniques , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Neovascularization, Pathologic/genetics , Neurofibromin 1/metabolism , Neurofibromin 1/physiology , Proteins/physiology , Signal Transduction/genetics , TOR Serine-Threonine Kinases , ras Proteins/metabolism
19.
J Neurosci ; 32(41): 14087-93, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23055477

ABSTRACT

Neurofibromatosis type 1 (NF1) is the most common monogenic disorder in which individuals manifest CNS abnormalities. Affected individuals develop glial neoplasms (optic gliomas, malignant astrocytomas) and neuronal dysfunction (learning disabilities, attention deficits). Nf1 genetically engineered mouse models have revealed the molecular and cellular underpinnings of gliomagenesis, attention deficit, and learning problems with relevance to basic neurobiology. Using NF1 as a model system, these studies have revealed critical roles for the NF1 gene in non-neoplastic cells in the tumor microenvironment, the importance of brain region heterogeneity, novel mechanisms of glial growth regulation, the neurochemical bases for attention deficit and learning abnormalities, and new insights into neural stem cell function. Here we review recent studies, presented at a symposium at the 2012 Society for Neuroscience annual meeting, that highlight unexpected cell biology insights into RAS and cAMP pathway effects on neural progenitor signaling, neuronal function, and oligodendrocyte lineage differentiation.


Subject(s)
Central Nervous System Diseases/metabolism , Disease Models, Animal , Neurofibromatosis 1/metabolism , Neurofibromin 1/physiology , Animals , Central Nervous System Diseases/genetics , Central Nervous System Diseases/physiopathology , Humans , Neurofibromatosis 1/genetics , Neurofibromatosis 1/physiopathology , Neurofibromin 1/genetics , Signal Transduction/physiology
20.
J Clin Invest ; 122(10): 3742-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945628

ABSTRACT

Intrinsic stress response pathways are frequently mobilized within tumor cells. The mediators of these adaptive mechanisms and how they contribute to carcinogenesis remain poorly understood. A striking example is heat shock factor 1 (HSF1), master transcriptional regulator of the heat shock response. Surprisingly, we found that loss of the tumor suppressor gene neurofibromatosis type 1 (Nf1) increased HSF1 levels and triggered its activation in mouse embryonic fibroblasts. As a consequence, Nf1-/- cells acquired tolerance to proteotoxic stress. This activation of HSF1 depended on dysregulated MAPK signaling. HSF1, in turn, supported MAPK signaling. In mice, Hsf1 deficiency impeded NF1-associated carcinogenesis by attenuating oncogenic RAS/MAPK signaling. In cell lines from human malignant peripheral nerve sheath tumors (MPNSTs) driven by NF1 loss, HSF1 was overexpressed and activated, which was required for tumor cell viability. In surgical resections of human MPNSTs, HSF1 was overexpressed, translocated to the nucleus, and phosphorylated. These findings reveal a surprising biological consequence of NF1 deficiency: activation of HSF1 and ensuing addiction to this master regulator of the heat shock response. The loss of NF1 function engages an evolutionarily conserved cellular survival mechanism that ultimately impairs survival of the whole organism by facilitating carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/physiology , Genes, Neurofibromatosis 1 , Neoplasm Proteins/physiology , Neurofibromin 1/deficiency , Transcription Factors/physiology , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heat Shock Transcription Factors , Hot Temperature , Humans , Leupeptins/toxicity , MAP Kinase Signaling System , Macrolides/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Nerve Sheath Neoplasms/pathology , Neurofibromin 1/physiology , Phosphorylation , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics , Withanolides/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...