Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Oncogene ; 36(12): 1669-1677, 2017 03 23.
Article in English | MEDLINE | ID: mdl-27748759

ABSTRACT

Neurofibromatosis type 1 (NF1) is an inherited disease in which affected patients are predisposed to develop benign Schwann cell (SC) tumours called neurofibromas. In the mouse, loss of Nf1 in the SC lineage causes neurofibroma formation. The tyrosine kinase receptor EGFR is expressed in Schwann cell precursors (SCP), which have been implicated in plexiform neurofibroma initiation. To test if EGFR activity affects neurofibroma initiation, size, and/or number, we studied mice expressing human EGFR in SCs and SCP in the context of mice that form neurofibromas. Neurofibroma number increased in homozygous CNP-hEGFR mice versus heterozygous littermates, and neurofibroma number and size increased when CNP-hEGFR was crossed to Nf1fl/fl;DhhCre mice. Conversely, diminished EGFR signalling in Nf1fl/fl;DhhCre;Wa2/+ mice decreased neurofibroma number. In vivo transplantation verified the correlation between EGFR activity and neurofibroma formation. Mechanistically, expression of CNP-hEGFR increased SCP/neurofibroma-initiating cell self-renewal, a surrogate for tumour initiation, and activated P-Stat3. Further, Il-6 reinforced Jak2/Stat3 activation in SCPs and SCs. These gain- and loss-of function assays show that levels of tyrosine kinase expression in SCPs modify neurofibroma initiation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Neurofibroma/metabolism , Neuroglia/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/genetics , Female , Homozygote , Humans , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Male , Mice , Mice, Transgenic , Mutation , Neurofibroma/mortality , Neurofibroma/pathology , Neurofibromin 1/genetics , Neuroglia/pathology , Tumor Burden
2.
Oncogene ; 33(49): 5626-36, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-24509877

ABSTRACT

Mutations in the neurofibromatosis type 1 (NF1) tumor suppressor gene are common in cancer and can cause resistance to therapy. Using transcriptome analysis we identified MAF as an NF1- regulated transcription factor and verified MAF regulation through RAS/MAPK/AP-1 signaling in malignant peripheral nerve sheath tumor (MPNST) cell lines. MAF was also downregulated in human MPNST. Acute re-expression of MAF promoted expression of glial differentiation markers in MPNST cells in vitro, decreased self-renewal of embryonic precursors and transiently affected tumor cell phenotypes in vitro by increasing MPNST cell death and reducing metabolic activity and anchorage-independent growth. Paradoxically, chronic MAF overexpression enhanced MPNST cell tumor growth in vivo, correlating with elevated pS6 in vitro and in vivo. RAD001 blocked MAF-mediated tumor growth, and MAF regulated the mTOR pathway through DEPTOR. MAPK inhibition with NF1 loss of function is predicted to show limited efficacy due to reactivation of mTOR signaling via MAF.


Subject(s)
MAP Kinase Signaling System , Nerve Sheath Neoplasms/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/metabolism , Proto-Oncogene Proteins c-maf/physiology , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Differentiation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Genes, Neurofibromatosis 1 , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Sheath Neoplasms/metabolism , Neurofibromatosis 1/metabolism , Neuroglia/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Schwann Cells/cytology , Signal Transduction , Transcriptome
3.
Oncogene ; 33(2): 173-80, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-23318430

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) develop sporadically or in the context of neurofibromatosis type 1. Epidermal growth factor receptor (EGFR) overexpression has been implicated in MPNST formation, but its precise role and relevant signaling pathways remain unknown. We found that EGFR overexpression promotes mouse neurofibroma transformation to aggressive MPNST (GEM-PNST). Immunohistochemistry demonstrated phosphorylated STAT3 (Tyr705) in both human MPNST and mouse GEM-PNST. A specific JAK2/STAT3 inhibitor FLLL32 delayed MPNST formation in an MPNST xenograft nude mouse model. STAT3 knockdown by shRNA prevented MPNST formation in vivo. Finally, reducing EGFR activity strongly reduced pSTAT3 in vivo. Thus, an EGFR-STAT3 pathway is necessary for MPNST transformation and establishment of MPNST xenografts growth but not for tumor maintenance. Efficacy of the FLLL32 pharmacological inhibitor in delaying MPNST growth suggests that combination therapies targeting JAK/STAT3 might be useful therapeutics.


Subject(s)
ErbB Receptors/physiology , Nerve Sheath Neoplasms/etiology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Curcumin/analogs & derivatives , Curcumin/pharmacology , Genes, Neurofibromatosis 1 , Humans , Janus Kinase 2/physiology , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/antagonists & inhibitors , Sarcoma/etiology
4.
Oncogene ; 32(9): 1135-43, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-22525268

ABSTRACT

Neurofibromatosis type 2 patients develop schwannomas, meningiomas and ependymomas resulting from mutations in the tumor suppressor gene, NF2, encoding a membrane-cytoskeleton adapter protein called merlin. Merlin regulates contact inhibition of growth and controls the availability of growth factor receptors at the cell surface. We tested if microtubule-based vesicular trafficking might be a mechanism by which merlin acts. We found that schwannoma cells, containing merlin mutations and constitutive activation of the Rho/Rac family of GTPases, had decreased intracellular vesicular trafficking relative to normal human Schwann cells. In Nf2-/- mouse Schwann (SC4) cells, re-expression of merlin as well as inhibition of Rac or its effector kinases, MLK and p38(SAPK), each increased the velocity of Rab6 positive exocytic vesicles. Conversely, an activated Rac mutant decreased Rab6 vesicle velocity. Vesicle motility assays in isolated squid axoplasm further demonstrated that both mutant merlin and active Rac specifically reduce anterograde microtubule-based transport of vesicles dependent upon the activity of p38(SAPK) kinase. Taken together, our data suggest loss of merlin results in the Rac-dependent decrease of anterograde trafficking of exocytic vesicles, representing a possible mechanism controlling the concentration of growth factor receptors at the cell surface.


Subject(s)
Genes, Neurofibromatosis 2/physiology , MAP Kinase Kinase Kinases/metabolism , Microtubules/metabolism , Neurofibromin 2/genetics , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Decapodiformes , Humans , Mutation , Neurilemmoma/metabolism , Neurofibromin 2/metabolism
5.
Oncogene ; 29(17): 2540-9, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20154721

ABSTRACT

The neurofibromatosis type 2 (NF2) tumor suppressor gene encodes merlin, a membrane/cytoskeleton protein necessary for the maintenance of contact inhibition of growth in cells. Bi-allelic inactivation of NF2 is known to cause multiple cancers in both humans and mice. However, the mechanism through which merlin exerts its tumor-suppressive function remains obscure. In this report, we show that NF2 knockout mouse embryonic fibroblasts lost contact inhibition of cell proliferation and contained significantly increased canonical Wnt signaling. Inhibition of Rac1, the activity of which is inversely regulated by NF2, through the use of a dominant-negative mutant, small hairpin RNA or a small molecule inhibitor in NF2-deficient cells, was able to suppress elevated Wnt signals as shown by reduced activity of the T-cell factor 4 (TCF4) transcription factor. Dominant-negative TCF4 or Rac1 mutant, as well as a small molecule inhibition of Wnt, were able to curb NF2 deficiency-elicited cell proliferation at the confluent state. Thus, Rac1-mediated canonical Wnt signaling is essential for the loss of contact inhibition in NF2-deficient cells.


Subject(s)
Contact Inhibition , Neurofibromin 2/physiology , Neuropeptides/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , rac GTP-Binding Proteins/physiology , Animals , Cell Proliferation , Cells, Cultured , Mice , Neurofibromin 2/deficiency , rac1 GTP-Binding Protein
6.
Oncogene ; 29(3): 368-79, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19901965

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas without effective therapeutics. Bioinformatics was used to identify potential therapeutic targets. Paired Box (PAX), Eyes Absent (EYA), Dachsund (DACH) and Sine Oculis (SIX) genes, which form a regulatory interactive network in Drosophila, were found to be dysregulated in human MPNST cell lines and solid tumors. We identified a decrease in DACH1 expression, and increases in the expressions of PAX6, EYA1, EYA2, EYA4, and SIX1-4 genes. Consistent with the observation that half of MPNSTs develop in neurofibromatosis type 1 (NF1) patients, subsequent to NF1 mutation, we found that exogenous expression of the NF1-GTPase activating protein-related domain normalized DACH1 expression. EYA4 mRNA was elevated more than 100-fold as estimated by quantitative real-time PCR in most MPNST cell lines. In vitro, suppression of EYA4 expression using short hairpin RNA reduced cell adhesion and migration and caused cellular necrosis without affecting cell proliferation or apoptotic cell death. MPNST cells expressing shEYA4 either failed to form tumors in nude mice or formed very small tumors, with extensive necrosis but similar levels of proliferation and apoptosis as control cells. Our findings identify a role of EYA4 and possibly interacting SIX and DACH proteins in MPNSTs and suggest the EYA4 pathway as a rational therapeutic target.


Subject(s)
Neoplasms, Experimental/genetics , Nerve Sheath Neoplasms/genetics , RNA Interference , Trans-Activators/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Necrosis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
7.
Neurology ; 69(18): 1761-71, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17967992

ABSTRACT

OBJECTIVE: We investigated the relationship between partial epilepsy, MRI findings, and atypical language representation. METHODS: A total of 102 patients (4 to 55 years) with left hemisphere epileptogenic zones were evaluated using three fMRI language tasks obtained at 1.5 or 3T with EPI BOLD techniques: verbal fluency, reading comprehension, and auditory comprehension. fMRI maps were visually interpreted at a standard threshold and rated as left or atypical language. RESULTS: Atypical language dominance occurred in 30 patients (29%) and varied with MRI type (p < 0.01). Atypical language representation occurred in 36% (13/36) with normal MRI, 21% (6/29) with mesial temporal sclerosis, 14% (4/28) with focal cortical lesions (dysplasia, tumor, vascular malformation), and all (6/6) with a history of stroke. Multivariate logistic regression analysis found handedness, seizure onset, and MRI type accounted for much of the variance in language activation patterns (chi(2) = 24.09, p < 0.01). Atypical language was more prevalent in patients with early seizure onset (43.2%, p < 0.05) and atypical handedness (60%, p < 0.01). None of the three clinical factors were correlated with each other (p > 0.40). Patients with atypical language had lower verbal abilities (F = 6.96, p = 0.01) and a trend toward lower nonverbal abilities (F = 3.58, p = 0.06). There were no differences in rates of atypical language across time, age groups, or MRI scanner. CONCLUSION: Early seizure onset and atypical handedness, as well as the location and nature of pathologic substrate, are important factors in language reorganization.


Subject(s)
Epilepsy, Complex Partial , Language Disorders , Verbal Behavior/physiology , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Epilepsy, Complex Partial/complications , Epilepsy, Complex Partial/physiopathology , Female , Functional Laterality , Humans , Infant , Intelligence Tests , Language Disorders/etiology , Language Disorders/physiopathology , Language Tests , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Retrospective Studies , Semantics
8.
Dev Cell ; 1(1): 63-72, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11703924

ABSTRACT

Mutations in the neurofibromatosis type II (NF2) tumor suppressor predispose humans and mice to tumor development. The study of Nf2+/- mice has demonstrated an additional effect of Nf2 loss on tumor metastasis. The NF2-encoded protein, merlin, belongs to the ERM (ezrin, radixin, and moesin) family of cytoskeleton:membrane linkers. However, the molecular basis for the tumor- and metastasis- suppressing activity of merlin is unknown. We have now placed merlin in a signaling pathway downstream of the small GTPase Rac. Expression of activated Rac induces phosphorylation and decreased association of merlin with the cytoskeleton. Furthermore, merlin overexpression inhibits Rac-induced signaling in a phosphorylation-dependent manner. Finally, Nf2-/- cells exhibit characteristics of cells expressing activated alleles of Rac. These studies provide insight into the normal cellular function of merlin and how Nf2 mutation contributes to tumor initiation and progression.


Subject(s)
Neurofibromin 2/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Gene Expression/physiology , Mice , Molecular Sequence Data , Neurofibromin 2/genetics , Phosphorylation , cdc42 GTP-Binding Protein/metabolism
9.
J Speech Lang Hear Res ; 44(5): 1116-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11708531

ABSTRACT

Many programs for the indirect management of stuttering in early childhood counsel adjustment of parental language models, which are presumed to play an exacerbating influence on vulnerable children's fluency. We examined the relative levels of linguistic demand in maternal language to stuttering and nonstuttering children, adjusted for each child's current level of linguistic development. No significant or observable differences were detected in the relative level of linguistic demand posed by parents of stuttering children very close to onset of symptoms. Empirical support for current advisement and potential ramifications are discussed.


Subject(s)
Language , Parent-Child Relations , Parents , Stuttering/prevention & control , Verbal Behavior , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Severity of Illness Index , Stuttering/diagnosis , Vocabulary
12.
Oncogene ; 20(1): 97-105, 2001 Jan 04.
Article in English | MEDLINE | ID: mdl-11244508

ABSTRACT

Loss of the tumor suppressor gene NF1 in neurofibromatosis type 1 (NF1) contributes to the development of a variety of tumors, including malignant peripheral nerve sheath tumors (MPNST) and benign neurofibromas. Of the different cell types found in neurofibromas, Schwann cells usually provide between 40 and 80%, and are thought to be critical for tumor growth. Here we describe the identification of growth factors that are upregulated in NF1-/- mouse Schwann cells and are potential regulators of angiogenesis and cell growth. Basic fibroblast growth factor (FGF-2), platelet-derived growth factor (PDGF) and midkine (MK) were found to be induced by loss of neurofibromin and MK was further characterized. MK was induced in human neurofibromas, schwannomas, and various nervous system tumors associated with NF1 or NF2; midkine showed an expression pattern overlapping but distinct from its homolog pleiotrophin (PTN). Immunohistochemistry revealed expression of MK in S-100 positive Schwann cells of dermal and plexiform neurofibromas, and in endothelial cells of tumor blood vessels, but not in normal blood vessels. Furthermore, MK demonstrated potent mitogenic activity for human systemic and brain endothelial cells in vitro and stimulated proliferation and soft agar colony formation of human MPNST derived S100 positive cells and fibroblastoid cells derived from an NF1 neurofibroma. The data support a possible central role for MK as a mediator of angiogenesis and neurofibroma growth in NF1. Oncogene (2001) 20, 97 - 105.


Subject(s)
Angiogenesis Inducing Agents/biosynthesis , Carrier Proteins/biosynthesis , Cytokines , Gene Expression Regulation, Neoplastic , Genes, Neurofibromatosis 1 , Mitogens/physiology , Nerve Tissue Proteins/deficiency , Neurofibroma/genetics , Schwann Cells/metabolism , Adult , Angiogenesis Inducing Agents/genetics , Angiogenesis Inducing Agents/physiology , Animals , Brain/blood supply , Brain/cytology , Brain/metabolism , Brain/pathology , Brain Neoplasms/chemistry , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Line , Endothelium, Vascular/chemistry , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Immunohistochemistry , Mice , Midkine , Nerve Tissue Proteins/genetics , Neurofibroma/metabolism , Neurofibroma/pathology , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromatosis 2/metabolism , Neurofibromin 1 , Schwann Cells/pathology , Tumor Cells, Cultured
13.
J Neurosci ; 21(4): 1110-6, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160381

ABSTRACT

In most mammalian cells, the cAMP-dependent protein kinase A pathway promotes growth arrest and cell differentiation. However in Schwann cells, the reverse is true. Elevated levels of cAMP function as the cofactor to a broad range of mitogenic cues in culture and in animals. Previous studies have suggested that cAMP acts at an early point in the Schwann cell mitogenic response, perhaps by stimulating the expression of growth factor receptors. We show here that cAMP acts downstream rather than upstream of growth factor receptor expression. The essential function(s) of cAMP is exerted as Schwann cells progress through the G(1) phase of the cell cycle. Ectopic expression studies using an inducible retroviral vector show that the G(1) phase requirement for cAMP can be alleviated by a single protein, cyclin D1. We show, in addition, that at least one function of the Nf1 tumor suppressor is to antagonize the accumulation of cAMP and the expression of cyclin D1 in Schwann cells. Thus a G(1) phase-specific protein, cyclin D1, accounts for two salient features of Schwann cell growth control: the promitotic response to cAMP and the antimitotic response to the Nf1 tumor suppressor.


Subject(s)
Cyclic AMP/metabolism , Cyclin D1/metabolism , Nerve Tissue Proteins/metabolism , Schwann Cells/metabolism , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/pharmacology , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Drug Synergism , Gene Expression/drug effects , Nerve Tissue Proteins/pharmacology , Neurofibromin 1 , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Promoter Regions, Genetic/drug effects , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Schwann Cells/cytology , Schwann Cells/drug effects , Tetracycline/pharmacology , Transfection
14.
J Neurosci Res ; 62(4): 491-502, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11070492

ABSTRACT

The product of the neurofibromatosis type II (NF2) tumor suppressor gene, merlin, is closely related to the ezrin-radixin-moesin (ERM) family, a group of proteins believed to link the cytoskeleton to the plasma membrane. Mutation in the NF2 locus is associated with Schwann cell tumors (schwannomas). The two predominant merlin isoforms, I and II, differ only in the carboxy-terminal 16 residues and only isoform I is anti-proliferative. Merlin lacks an actin-binding domain conserved among ezrin, radixin and moesin. Because merlin, ezrin and moesin are co-expressed in Schwann cells, and all homodimerize, we have examined whether merlin and ezrin dimerize with one another. We found by immunoprecipitation and yeast two-hybrid assays that both merlin isoforms interact with ezrin. The interaction occurs in a head-to-tail orientation, with the amino-terminal half of one protein interacting with the carboxy-terminal half of the other. The two merlin isoforms behave differently in their interaction with ezrin. Isoform I binds only ezrin whose carboxy-terminus is exposed, whereas isoform II binds ezrin regardless of whether ezrin is in the open or closed conformation. The heterodimerization of merlin is a much stronger interaction than the interaction between either merlin isoform and ezrin, and can inhibit merlin-ezrin binding. This suggests that, in vivo, merlin dimerization could regulate merlin-ERM protein interaction, and could thus indirectly regulate other interactions involving ERM proteins.


Subject(s)
Cytoskeletal Proteins/genetics , Genes, Tumor Suppressor/physiology , Membrane Proteins/genetics , Neurofibromatosis 2/genetics , Phosphoproteins/genetics , Protein Isoforms/genetics , Animals , Animals, Newborn , Blood Proteins/genetics , Blood Proteins/metabolism , Cytoskeletal Proteins/metabolism , Humans , Membrane Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/physiopathology , Neurofibromin 2 , Phosphoproteins/metabolism , Protein Isoforms/metabolism , Protein Structure, Tertiary/physiology , RNA, Messenger/metabolism , Rats , Schwann Cells/metabolism , Schwann Cells/pathology , Tumor Cells, Cultured
15.
J Speech Lang Hear Res ; 43(5): 1252-63, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11063245

ABSTRACT

There has been clinical speculation that parents of young stuttering children have expectations of their children's communication abilities that are not well-matched to the children's actual skills. We appraised the language abilities of 15 children close to the onset of stuttering symptoms and 15 age-, sex-, and SES-matched fluent children using an array of standardized tests and spontaneous language sample measures. Parents concurrently completed two parent-report measures of the children's communicative development. Results indicated generally depressed performance on all child speech and language measures by the children who stutter. Parent report was closely attuned to child performance for the stuttering children; parents of nonstuttering children were less accurate in their predictions of children's communicative performance. Implications for clinical advisement to parents of stuttering children are discussed.


Subject(s)
Child Language , Communication , Parents , Speech Perception , Stuttering/psychology , Verbal Behavior , Child, Preschool , Counseling , Female , Humans , Infant , Male , Severity of Illness Index , Stuttering/diagnosis
16.
J Neurogenet ; 14(2): 63-106, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10992163
17.
J Cell Biol ; 150(5): 1071-84, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10973996

ABSTRACT

We describe a key role for the CD44 transmembrane glycoprotein in Schwann cell-neuron interactions. CD44 proteins have been implicated in cell adhesion and in the presentation of growth factors to high affinity receptors. We observed high CD44 expression in early rat neonatal nerves at times when Schwann cells proliferate but low expression in adult nerves, where CD44 was found in some nonmyelinating Schwann cells and to varying extents in some myelinating fibers. CD44 constitutively associated with erbB2 and erbB3, receptor tyrosine kinases that heterodimerize and signal in Schwann cells in response to neuregulins. Moreover, CD44 significantly enhanced neuregulin-induced erbB2 phosphorylation and erbB2-erbB3 heterodimerization. Reduction of CD44 expression in vitro resulted in loss of Schwann cell-neurite adhesion and Schwann cell apoptosis. CD44 is therefore crucial for maintaining neuron-Schwann cell interactions at least partly by facilitating neuregulin-induced erbB2-erbB3 activation.


Subject(s)
Hyaluronan Receptors/physiology , Neuregulin-1/physiology , Neurons/physiology , Schwann Cells/physiology , Animals , Animals, Newborn , Cell Adhesion , Cell Communication , Cells, Cultured , Coculture Techniques , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Humans , Models, Neurological , Neurites/physiology , Neurons/cytology , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Schwann Cells/cytology , Sciatic Nerve/cytology , Sciatic Nerve/physiology , Signal Transduction
18.
J Biol Chem ; 275(39): 30740-5, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-10900196

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by multiple neurofibromas, peripheral nerve tumors containing mainly Schwann cells and fibroblasts. The NF1 gene encodes neurofibromin, a tumor suppressor postulated to function in part as a Ras GTPase-activating protein. The roles of different cell types and of elevated Ras-GTP in neurofibroma formation are unclear. To determine which neurofibroma cell type has altered Ras-GTP regulation, we developed an immunocytochemical assay for active, GTP-bound Ras. In NIH 3T3 cells, the assay detected overexpressed, constitutively activated K-, N-, and Ha-Ras and insulin-induced endogenous Ras-GTP. In dissociated neurofibroma cells from NF1 patients, Ras-GTP was elevated in Schwann cells but not fibroblasts. Twelve to 62% of tumor Schwann cells showed elevated Ras-GTP, unexpectedly revealing neurofibroma Schwann cell heterogeneity. Increased basal Ras-GTP did not correlate with increased cell proliferation. Normal human Schwann cells, however, did not demonstrate elevated basal Ras activity. Furthermore, compared with cells from wild type littermates, Ras-GTP was elevated in all mouse Nf1(-/-) Schwann cells but never in Nf1(-/-) mouse fibroblasts. Our results indicate that Ras activity is detectably increased in only some neurofibroma Schwann cells and suggest that neurofibromin is not an essential regulator of Ras activity in fibroblasts.


Subject(s)
Fibroblasts/chemistry , Guanosine Triphosphate/isolation & purification , Histocytochemistry/methods , Neurofibroma/chemistry , Schwann Cells/chemistry , ras Proteins/isolation & purification , Animals , Cell Separation , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Neurofibroma/pathology , Neurofibromin 1 , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , ras GTPase-Activating Proteins/genetics
19.
J Invest Dermatol ; 114(6): 1093-100, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10844550

ABSTRACT

There is increasing evidence implicating the human NF1 gene in epithelial carcinogenesis. To test if NF1 can play a part in skin tumor formation, we analyzed effects of the skin cancer initiator dimethylbenz-anthracene and/or the tumor promoter 12-O-tetradecanoyl-13-acetylphorbol on mice heterozygous for null mutations in Nf1 (Nf1+/-). Mice were on the C57BL/6 background, noted for resistance to chemical carcinogens. Nf1+/- mice (18 of 24) developed papillomas after treatment with dimethylbenzanthracene and 12-O-tetradecanoyl-13-acetylphorbol; papillomas did not develop in wild-type C57BL/6 mice nor Nf1+/- mice treated with 12-O-tetradecanoyl-13-acetylphorbol alone. All papillomas analyzed (six of six) had mutations in codon 61 of H-ras, demonstrating strong cooperation between the Nf1 GTPase activating protein for Ras, neurofibromin, and Ras-GTP. After exposure to 12-O-tetradecanoyl-13-acetylphorbol, Nf1+/- keratinocytes showed significant, sustained, increases in proliferation, implicating Nf1 in phorbol ester responsive pathways. Thus, Nf1 levels regulate the response of keratinocytes to 12-O-tetradecanoyl-13-acetylphorbol. Nf1+/- mice also showed a 2-fold increase in the development of pigmented skin patches stimulated by dimethylbenzanthracene; patches were characterized by hair follicles in anagen phase, implicating keratinocytes in the aberrant hyperpigmentation. Our results show that mutation in the Nf1 gene causes abnormal keratinocyte proliferation that can be revealed by environmental assaults such as carcinogen exposure. The data support a plausible role for NF1 mutation in human epithelial carcinogenesis.


Subject(s)
Carcinogens/pharmacology , Genes, Neurofibromatosis 1/physiology , Mice, Inbred C57BL/genetics , Papilloma/metabolism , Skin Neoplasms/metabolism , Skin Pigmentation/drug effects , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , DNA Mutational Analysis , Female , Genes, ras/genetics , Humans , Mice , Papilloma/genetics , Skin/drug effects , Skin Neoplasms/genetics , Skin Pigmentation/genetics , Tetradecanoylphorbol Acetate/pharmacology
20.
J Clin Invest ; 105(9): 1233-41, 2000 May.
Article in English | MEDLINE | ID: mdl-10791998

ABSTRACT

We have found that EGF-R expression is associated with the development of the Schwann cell-derived tumors characteristic of neurofibromatosis type 1 (NF1) and in animal models of this disease. This is surprising, because Schwann cells normally lack EGF-R and respond to ligands other than EGF. Nevertheless, immunoblotting, Northern analysis, and immunohistochemistry revealed that each of 3 malignant peripheral nerve sheath tumor (MPNST) cell lines from NF1 patients expressed the EGF-R, as did 7 of 7 other primary MPNSTs, a non-NF1 MPNST cell line, and the S100(+) cells from each of 9 benign neurofibromas. Furthermore, transformed derivatives of Schwann cells from NF1(-/-) mouse embryos also expressed the EGF-R. All of the cells or cell lines expressing EGF-R responded to EGF by activation of downstream signaling pathways. Thus, EGF-R expression may play an important role in NF1 tumorigenesis and Schwann cell transformation. Consistent with this hypothesis, growth of NF1 MPNST lines and the transformed NF1(-/-) mouse embryo Schwann cells was greatly stimulated by EGF in vitro and could be blocked by agents that antagonize EGF-R function.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Neurofibromatosis 1/metabolism , Proteins/genetics , Animals , Cell Transformation, Neoplastic , Humans , Mice , Mice, Mutant Strains , Neurilemmoma , Neurofibromin 1 , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL