Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neuropathol Exp Neurol ; 74(6): 568-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25946318

ABSTRACT

Neurofibromin, the tumor suppressor encoded by the neurofibromatosis type 1 (NF1) gene, potentially suppresses the activation of H-Ras, N-Ras, and K-Ras. However, it is not known whether these classic Ras proteins are hyperactivated in NF1-null nerve sheath tumors, how they contribute to tumorigenesis, and what signaling pathways mediate their effects. Here we show that H-Ras, N-Ras, and K-Ras are coexpressed with their activators (guanine nucleotide exchange factors) in neurofibromin-null malignant peripheral nerve sheath tumor (MPNST) cells, and that all 3 Ras proteins are activated. Dominant negative (DN) H-Ras, a pan-inhibitor of the classic Ras family, inhibited MPNST proliferation and survival, but not migration. However, NF1-null MPNST cells were variably dependent on individual Ras proteins. In some lines, ablation of H-Ras, N-Ras, and/or K-Ras inhibited mitogenesis. In others, ablation of a single Ras protein had no effect on proliferation; in these lines, ablation of a single Ras protein resulted in compensatory increases in the activation and/or expression of other Ras proteins. Using mass spectrometry-based phosphoproteomics, we identified 7 signaling networks affecting morphology, proliferation, and survival that are regulated by DN H-Ras. Thus, neurofibromin loss activates multiple classic Ras proteins that promote proliferation and survival by regulating several distinct signaling cascades.


Subject(s)
Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Neurofibromatosis 1/metabolism , ras Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Chromatography, Liquid , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation/genetics , Nerve Sheath Neoplasms/pathology , Neurofibromatosis 1/genetics , Phosphoproteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Tandem Mass Spectrometry , Transfection , ras Proteins/genetics
2.
J Neuropathol Exp Neurol ; 73(11): 1078-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25289889

ABSTRACT

Chemotherapeutic agents effective against malignant peripheral nerve sheath tumors (MPNSTs) are urgently needed. We recently found that tamoxifen potently impedes xenograft growth. In vitro, tamoxifen inhibits MPNST proliferation and survival in an estrogen receptor-independent manner; these effects are phenocopied by the calmodulin inhibitor trifluoperazine. The present study was performed to establish the mechanism of action of tamoxifen in vivo and optimize its therapeutic effectiveness. To determine if tamoxifen has estrogen receptor-dependent effects in vivo, we grafted MPNST cells in castrated and ovariectomized mice; xenograft growth was unaffected by reductions in sex hormones. To establish whether tamoxifen and trifluoperazine additively or synergistically impede MPNST growth, mice xenografted with neurofibromatosis type 1-associated or sporadic MPNST cells were treated with tamoxifen, trifluoperazine, or both drugs for 30 days. Both monotherapies inhibited graft growth by 50%, whereas combinatorial treatment maximally reduced graft mass by 90% and enhanced decreases in proliferation and survival. Kinomic analyses showed that tamoxifen and trifluoperazine have both shared and distinct targets in MPNSTs. In addition, trifluoperazine prevented tamoxifen-induced increases in serum/glucocorticoid regulated kinase 1, a protein linked to tamoxifen resistance. These findings suggest that combinatorial therapy with tamoxifen and trifluoperazine is effective against MPNSTs because these agents target complementary pathways that are essential for MPNST pathogenesis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems/methods , Neurilemmoma/drug therapy , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Female , Male , Mice , Neurilemmoma/pathology , Signal Transduction/physiology , Tamoxifen/administration & dosage , Treatment Outcome , Trifluoperazine/administration & dosage , Xenograft Model Antitumor Assays/methods
3.
Acta Neuropathol ; 127(4): 573-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24232507

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are Schwann cell-derived malignancies that arise from plexiform neurofibromas in patients with mutation of the neurofibromin 1 (NF1) gene. We have shown that the growth factor neuregulin-1 (NRG1) also contributes to human neurofibroma and MPNST pathogenesis and that outbred C57BL/6J × SJL/J transgenic mice overexpressing NRG1 in Schwann cells (P0-GGFß3 mice) recapitulate the process of neurofibroma-MPNST progression. However, it is unclear whether NRG1 acts predominantly within NF1-regulated signaling cascades or instead activates other essential cascades that cooperate with NF1 loss to promote tumorigenesis. We now report that tumorigenesis is suppressed in inbred P0-GGFß3 mice on a C57BL/6J background. To determine whether NRG1 overexpression interacts with reduced Nf1 or Trp53 gene dosage to "unmask" tumorigenesis in these animals, we followed cohorts of inbred P0-GGFß3;Nf1+/−, P0-GGFß3;Trp53+/− and control (P0-GGFß3, Nf1+/− and Trp53+/−) mice for 1 year. We found no reduction in survival or tumors in control and P0-GGFß3;Nf1+/− mice. In contrast, P0-GGFß3;Trp53+/− mice died on average at 226 days, with MPNSTs present in 95 % of these mice. MPNSTs in inbred P0-GGFß3;Trp53+/− mice arose de novo from micro-MPNSTs that uniformly develop intraganglionically. These micro-MPNSTs are of lower grade (WHO grade II-III) than the major MPNSTs (WHO grade III-IV); array comparative genomic hybridization showed that lower grade MPNSTs also had fewer genomic abnormalities. Thus, P0-GGFß3;Trp53+/− mice represent a novel model of low- to high-grade MPNST progression. We further conclude that NRG1 promotes peripheral nervous system neoplasia predominantly via its effects on the signaling cascades affected by Nf1 loss.


Subject(s)
Gene Expression , Haploinsufficiency/genetics , Neuregulin-1/metabolism , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Desmin/metabolism , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Myelin P0 Protein/genetics , Myelin P0 Protein/metabolism , Neurofibroma/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology
4.
Cancer Res ; 73(14): 4395-405, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23722551

ABSTRACT

Tamoxifen is widely used to treat estrogen receptor-positive breast cancer. Recent findings that tamoxifen and its derivative 4-hydroxytamoxifen (OHT) can exert estrogen receptor-independent cytotoxic effects have prompted the initiation of clinical trials to evaluate its use in estrogen receptor-negative malignancies. For example, tamoxifen and OHT exert cytotoxic effects in malignant peripheral nerve sheath tumors (MPNST) where estrogen is not involved. In this study, we gained insights into the estrogen receptor-independent cytotoxic effects of OHT by studying how it kills MPNST cells. Although caspases were activated following OHT treatment, caspase inhibition provided no protection from OHT-induced death. Rather, OHT-induced death in MPNST cells was associated with autophagic induction and attenuated by genetic inhibition of autophagic vacuole formation. Mechanistic investigations revealed that OHT stimulated autophagic degradation of K-Ras, which is critical for survival of MPNST cells. Similarly, we found that OHT induced K-Ras degradation in breast, colon, glioma, and pancreatic cancer cells. Our findings describe a novel mechanism of autophagic death triggered by OHT in tumor cells that may be more broadly useful clinically in cancer treatment.


Subject(s)
Autophagy/drug effects , Cell Death/drug effects , Nerve Sheath Neoplasms/drug therapy , Proto-Oncogene Proteins/metabolism , Tamoxifen/analogs & derivatives , ras Proteins/metabolism , Autophagy/genetics , Caspases/genetics , Caspases/metabolism , Cell Death/genetics , Cell Line, Tumor , Down-Regulation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Sheath Neoplasms/enzymology , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , ras Proteins/genetics
5.
J Neuropathol Exp Neurol ; 72(3): 219-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23399900

ABSTRACT

Aberrant epidermal growth factor receptor (EGFR) expression promotes the pathogenesis of malignant peripheral nerve sheath tumors (MPNSTs), the most common malignancy associated with neurofibromatosis type 1, but the mechanisms by which EGFR expression promotes MPNST pathogenesis are poorly understood. We hypothesized that inappropriately expressed EGFRs promote MPNST invasion and found that these kinases are concentrated in MPNST invadopodia in vitro. Epidermal growth factor receptor knockdown inhibited the migration of unstimulated MPNST cells in vitro, and exogenous EGF further enhanced MPNST migration in a substrate-specific manner, promoting migration on laminin and, to a lesser extent, collagen. In this setting, EGF acts as a chemotactic factor. We also found that the 7 known EGFR ligands (EGF, betacellulin, epiregulin, heparin-binding EGF, transforming growth factor-α [TGF-α], amphiregulin, and epigen) variably enhanced MPNST migration in a concentration-dependent manner, with TGF-α being particularly potent. With the exception of epigen, these factors similarly promoted the migration of nonneoplastic Schwann cells. Although transcripts encoding all 7 EGFR ligands were detected in human MPNST cells and tumor tissues, only TGF-α was consistently overexpressed and was found to colocalize with EGFR in situ. These data indicate that constitutive EGFR activation, potentially driven by autocrine or paracrine TGF-α signaling, promotes the aggressive invasive behavior characteristic of MPNSTs.


Subject(s)
ErbB Receptors/metabolism , Neoplasm Invasiveness/pathology , Nerve Sheath Neoplasms/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Gene Knockdown Techniques , Humans , Immunoblotting , Immunohistochemistry , Ligands , Nerve Sheath Neoplasms/pathology , Rats , Real-Time Polymerase Chain Reaction , Schwann Cells , Transforming Growth Factor alpha/metabolism
6.
Am J Pathol ; 182(3): 646-67, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23321323

ABSTRACT

Patients with neurofibromatosis type 1 (NF1) develop benign plexiform neurofibromas that frequently progress to become malignant peripheral nerve sheath tumors (MPNSTs). A genetically engineered mouse model that accurately models plexiform neurofibroma-MPNST progression in humans would facilitate identification of somatic mutations driving this process. We previously reported that transgenic mice overexpressing the growth factor neuregulin-1 in Schwann cells (P(0)-GGFß3 mice) develop MPNSTs. To determine whether P(0)-GGFß3 mice accurately model human neurofibroma-MPNST progression, cohorts of these animals were monitored through death and were necropsied; 94% developed multiple neurofibromas, with 70% carrying smaller numbers of MPNSTs. Nascent MPNSTs were identified within neurofibromas, suggesting that these sarcomas arise from neurofibromas. Although neurofibromin expression was maintained, P(0)-GGFß3 MPNSTs exhibited Ras hyperactivation, as in human NF1-associated MPNSTs. P(0)-GGFß3 MPNSTs also exhibited abnormalities in the p16(INK4A)-cyclin D/CDK4-Rb and p19(ARF)-Mdm-p53 pathways, analogous to their human counterparts. Array comparative genomic hybridization (CGH) demonstrated reproducible chromosomal alterations in P(0)-GGFß3 MPNST cells (including universal chromosome 11 gains) and focal gains and losses affecting 39 neoplasia-associated genes (including Pten, Tpd52, Myc, Gli1, Xiap, and Bbc3/PUMA). Array comparative genomic hybridization also identified recurrent focal copy number variations affecting genes not previously linked to neurofibroma or MPNST pathogenesis. We conclude that P(0)-GGFß3 mice represent a robust model of neurofibroma-MPNST progression useful for identifying novel genes driving neurofibroma and MPNST pathogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomes, Mammalian/genetics , DNA Copy Number Variations/genetics , Disease Progression , Nerve Sheath Neoplasms/pathology , Neuregulin-1/metabolism , Neurofibroma/pathology , Animals , Base Pairing/genetics , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Transformation, Neoplastic/pathology , Comparative Genomic Hybridization , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Sheath Neoplasms/genetics , Neurofibroma/genetics , Neurofibromin 1/metabolism , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Signal Transduction/genetics , ras Proteins/metabolism
7.
J Vis Exp ; (49)2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21460792

ABSTRACT

Although in vitro screens are essential for the initial identification of candidate therapeutic agents, a rigorous assessment of the drug's ability to inhibit tumor growth must be performed in a suitable animal model. The type of animal model that is best for this purpose is a topic of intense discussion. Some evidence indicates that preclinical trials examining drug effects on tumors arising in transgenic mice are more predictive of clinical outcome(1)and so candidate therapeutic agents are often tested in these models. Unfortunately, transgenic models are not available for many tumor types. Further, transgenic models often have other limitations such as concerns as to how well the mouse tumor models its human counterpart, incomplete penetrance of the tumor phenotype and an inability to predict when tumors will develop. Consequently, many investigators use xenograft models (human tumor cells grafted into immunodeficient mice) for preclinical trials if appropriate transgenic tumor models are not available. Even if transgenic models are available, they are often partnered with xenograft models as the latter facilitate rapid determination of therapeutic ranges. Further, this partnership allows a comparison of the effectiveness of the agent in transgenic tumors and genuine human tumor cells. Historically, xenografting has often been performed by injecting tumor cells subcutaneously (ectopic xenografts). This technique is rapid and reproducible, relatively inexpensive and allows continuous quantitation of tumor growth during the therapeutic period(2). However, the subcutaneous space is not the normal microenvironment for most neoplasms and so results obtained with ectopic xenografting can be misleading due to factors such as an absence of organ-specific expression of host tissue and tumor genes. It has thus been strongly recommended that ectopic grafting studies be replaced or complemented by studies in which human tumor cells are grafted into their tissue of origin (orthotopic xenografting)(2). Unfortunately, implementation of this recommendation is often thwarted by the fact that orthotopic xenografting methodologies have not yet been developed for many tumor types. Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive sarcomas that occur sporadically or in association with neurofibromatosis type 1(3) and most commonly arise in the sciatic nerve(4). Here we describe a technically straightforward method in which firefly luciferase-tagged human MPNST cells are orthopically xenografted into the sciatic nerve of immunodeficient mice. Our approach to assessing the success of the grafting procedure in individual animals and subsequent non-biased randomization into study groups is also discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , Animals , Humans , Mice , Mice, Inbred NOD , Mice, SCID
8.
Neuro Oncol ; 13(1): 28-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21075781

ABSTRACT

Few therapeutic options are available for malignant peripheral nerve sheath tumors (MPNSTs), the most common malignancy associated with neurofibromatosis type 1 (NF1). Guided by clinical observations suggesting that some NF1-associated nerve sheath tumors are hormonally responsive, we hypothesized that the selective estrogen receptor (ER) modulator tamoxifen would inhibit MPNST tumorigenesis in vitro and in vivo. To test this hypothesis, we examined tamoxifen effects on MPNST cell proliferation and survival, MPNST xenograft growth, and the mechanism by which tamoxifen impeded these processes. We found that 1-5 µM 4-hydroxy-tamoxifen induced MPNST cell death, whereas 0.01-0.1 µM 4-hydroxy-tamoxifen inhibited mitogenesis. Dermal and plexiform neurofibromas, MPNSTs, and MPNST cell lines expressed ERß and G-protein-coupled ER-1 (GPER); MPNSTs also expressed estrogen biosynthetic enzymes. However, MPNST cells did not secrete 17ß-estradiol, exogenous 17ß-estradiol did not stimulate mitogenesis or rescue 4-hydroxy-tamoxifen effects on MPNST cells, and the steroidal antiestrogen ICI-182,780 did not mimic tamoxifen effects on MPNST cells. Further, ablation of ERß and GPER had no effect on MPNST proliferation, survival, or tamoxifen sensitivity, indicating that tamoxifen acts via an ER-independent mechanism. Consistent with this hypothesis, inhibitors of calmodulin (trifluoperazine, W-7), another known tamoxifen target, recapitulated 4-hydroxy-tamoxifen effects on MPNST cells. Tamoxifen was also effective in vivo, demonstrating potent antitumor activity in mice orthotopically xenografted with human MPNST cells. We conclude that 4-hydroxy-tamoxifen inhibits MPNST cell proliferation and survival via an ER-independent mechanism. The in vivo effectiveness of tamoxifen provides a rationale for clinical trials in cases of MPNSTs.


Subject(s)
Cell Proliferation/drug effects , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/metabolism , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Estrogens/metabolism , Humans , Mice , Nerve Sheath Neoplasms/pathology , Neurofibroma, Plexiform/drug therapy , Neurofibroma, Plexiform/metabolism , Neurofibroma, Plexiform/pathology , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tamoxifen/analogs & derivatives , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Glia ; 57(14): 1501-20, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19306381

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are the most common malignancy associated with neurofibromatosis Type 1 (NF1). These Schwann cell lineage-derived sarcomas aggressively invade adjacent nerve and soft tissue, frequently precluding surgical resection. Little is known regarding the mechanisms underlying this invasive behavior. We have shown that MPNSTs express neuregulin-1 (NRG-1) beta isoforms, which promote Schwann cell migration during development, and NRG-1 alpha isoforms, whose effects on Schwann cells are poorly understood. Hypothesizing that NRG-1 beta and/or NRG-1 alpha promote MPNST invasion, we found that NRG-1 beta promoted MPNST migration in a substrate-specific manner, markedly enhancing migration on laminin but not on collagen type I or fibronectin. The NRG-1 receptors erbB3 and erbB4 were present in MPNST invadopodia (processes mediating invasion), partially colocalized with focal adhesion kinase and the laminin receptor beta(1)-integrin and coimmunoprecipitated with beta(1)-integrin. NRG-1 beta stimulated human and murine MPNST cell migration and invasion in a concentration-dependent manner in three-dimensional migration assays, acting as a chemotactic factor. Both baseline and NRG-1 beta-induced migration were erbB-dependent and required the action of MEK 1/2, SAPK/JNK, PI-3 kinase, Src family kinases and ROCK-I/II. In contrast, NRG-1 alpha had no effect on the migration and invasion of some MPNST lines and inhibited the migration of others. While NRG-1 beta potently and persistently activated Erk 1/2, SAPK/JNK, Akt and Src family kinases, NRG-1 alpha did not activate Akt and activated these other kinases with kinetics distinct from those evident in NRG-1 beta-stimulated cells. These findings suggest that NRG-1 beta enhances MPNST migration and that NRG-1 beta and NRG-1 alpha differentially modulate this process.


Subject(s)
Cell Movement/physiology , Nerve Sheath Neoplasms/physiopathology , Neuregulin-1/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Chemotaxis/physiology , Collagen Type I/metabolism , ErbB Receptors/metabolism , Fibronectins/metabolism , Humans , Integrin beta1/metabolism , Kinetics , Laminin/metabolism , Mice , Mice, Inbred C57BL , Nerve Sheath Neoplasms/enzymology , Protein Isoforms/metabolism , Receptor, ErbB-3/metabolism , Receptor, ErbB-4 , Schwann Cells/physiology
10.
J Neuropathol Exp Neurol ; 65(2): 162-75, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462207

ABSTRACT

Sporadic and neurofibromatosis type 2-associated schwannomas contain a glial growth factor (GGF)-like activity that has been hypothesized to promote neoplastic Schwann cell mitogenesis. It is not known whether this GGF-like activity is neuregulin-1 (NRG-1), an epidermal growth factor (EGF)-related molecule that regulates the proliferation, survival, and differentiation of developing Schwann cells, the related factor NRG-2, or another NRG/EGF ligand. We report that neoplastic Schwann cells within schwannomas overexpress multiple alpha and beta transmembrane precursors from the class II and class III NRG-1 subfamilies. NRG-2 alpha and beta transcripts are similarly overexpressed in some tumors. Of the other 8 known NRG/EGF ligands, only heparin-binding EGF, epiregulin, and TGFalpha are detectable in schwannomas. Neoplastic Schwann cells almost uniformly express erbB2 and erbB3, 2 membrane receptor tyrosine kinases mediating NRG-1 and NRG-2 action. Expression of the NRG receptor erbB4 and EGF receptor is also evident in schwannomas, but is more limited, occurring in only a subset of these tumors. ErbB2, the preferred dimerization partner for all erbB kinases, is constitutively phosphorylated in schwannomas. These observations suggest that autocrine, paracrine, and/or juxtacrine NRG-1/NRG-2 signaling promotes schwannoma pathogenesis and that this signaling pathway may be an important therapeutic target in schwannomas.


Subject(s)
Epidermal Growth Factor/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurilemmoma , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Adult , Aged , Epidermal Growth Factor/genetics , Female , Humans , Male , Middle Aged , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Neuregulin-1 , Neurilemmoma/pathology , Neurilemmoma/physiopathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Oncogene ; 24(36): 5589-605, 2005 Aug 25.
Article in English | MEDLINE | ID: mdl-15897877

ABSTRACT

Patients with neurofibromatosis type 1 develop aggressive Schwann cell neoplasms known as malignant peripheral nerve sheath tumors (MPNSTs). Although tumor suppressor gene mutations play an important role in MPNST pathogenesis, it is likely that dysregulated signaling by as yet unidentified growth factors also contributes to the formation of these sarcomas. To test the hypothesis that neuregulin-1 (NRG-1) growth factors promote mitogenesis in MPNSTs, we examined the expression and action of NRG-1 in human MPNSTs and neurofibromas, the benign precursor lesions from which MPNSTs arise. Multiple alpha and beta transmembrane precursors from the class II and III NRG-1 subfamilies are present in both tumor types. Neoplastic Schwann cells within these neoplasms variably express the erbB kinases mediating NRG-1 responses (erbB2, erbB3 and/or erbB4). Human MPNST cell lines (Mash-1, YST-1, NMS-2 and NMS-2PC cells) similarly coexpress multiple NRG-1 isoforms and erbB receptors. These MPNST lines are NRG-1 responsive and demonstrate constitutive erbB phosphorylation. Treatment with PD168393 and PD158780, two structurally and mechanistically distinct erbB inhibitors, abolishes erbB phosphorylation and reduces DNA synthesis in these lines. These findings suggest that autocrine and/or paracrine NRG-1/erbB signaling promotes neoplastic Schwann cell proliferation and may be an important therapeutic target in neurofibromas and MPNSTs.


Subject(s)
Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Neuregulin-1/metabolism , Oncogene Proteins v-erbB/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Signal Transduction , Biomarkers , Cell Proliferation , DNA/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Male , Neuregulin-1/genetics , Oncogene Proteins v-erbB/antagonists & inhibitors , Phosphotyrosine/metabolism , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
12.
J Neuropathol Exp Neurol ; 63(11): 1144-54, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15581182

ABSTRACT

In both humans and animal models, diabetic sympathetic autonomic neuropathy is associated with the selective development of markedly enlarged distal axons and nerve terminals (neuroaxonal dystrophy, NAD). NAD occurs in the prevertebral superior mesenteric and celiac ganglia (SMG-CG), but not in the paravertebral superior cervical ganglion (SCG). To identify molecular differences between these ganglia that may explain their selective vulnerability to NAD, we have examined global gene expression patterns in control and diabetic rat sympathetic ganglia before and after the onset of structural evidence of NAD. As predicted, major differences in transcriptional profiles exist between SCG and SMG-CG in normal young adult animals including, but not limited to, known differences in neurotransmitter-related gene expression. Gene expression patterns of diabetic SMG-CG and SCG, prior to the development of NAD lesions, also differ from their age-matched non-diabetic counterparts. However, diabetes has ganglion-specific effects on gene expression; of approximately 110 transcripts that were differentially expressed between diabetic and control sympathetic ganglia, only 5 were differentially expressed as a result of diabetes in both SCG and SMG-CG. Genes involving synapse and mitochondrial structure and function, oxidative stress, and glycolysis were highly represented in the differentially expressed gene set. Differences in the number of synapse-related gene alterations in diabetic SMG-CG (18 genes) versus SCG (2 genes) prior to the onset of NAD may also well explain the selective development of NAD in the SMG-CG. These results provide support for the specificity of diabetes-modulated gene expression for selected neuronal subpopulations of sympathetic noradrenergic neurons.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Ganglia, Sympathetic/physiology , Gene Expression , Neuroaxonal Dystrophies/etiology , Animals , Diabetes Mellitus, Experimental/complications , Gene Expression Profiling , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Superior Cervical Ganglion/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...