Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
iScience ; 26(2): 106096, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36818284

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are soft-tissue sarcomas of the peripheral nervous system that develop either sporadically or in the context of neurofibromatosis type 1 (NF1). MPNST diagnosis can be challenging and treatment outcomes are poor. We present here a resource consisting of the genomic characterization of 9 widely used human MPNST cell lines for their use in translational research. NF1-related cell lines recapitulated primary MPNST copy number profiles, exhibited NF1, CDKN2A, and SUZ12/EED tumor suppressor gene (TSG) inactivation, and presented no gain-of-function mutations. In contrast, sporadic cell lines collectively displayed different TSG inactivation patterns and presented kinase-activating mutations, fusion genes, altered mutational frequencies and COSMIC signatures, and different methylome-based classifications. Cell lines re-classified as melanomas and other sarcomas exhibited a different drug-treatment response. Deep genomic analysis, methylome-based classification, and cell-identity marker expression, challenged the identity of common MPNST cell lines, opening an opportunity to revise MPNST differential diagnosis.

2.
J Neurochem ; 137(4): 659-60, 2016 May.
Article in English | MEDLINE | ID: mdl-27062176

ABSTRACT

This is an obituary for Richard Hudson Quarles, an internationally renowned neuroscientist, who retired in 2007 after 39 years at the National Institutes of Health, and who died August 9, 2015 in Sandy Spring, Maryland, USA. Richard Hudson Quarles, circa 1984, courtesy of The NIH Record newsletter.


Subject(s)
Medical Laboratory Personnel/history , National Institutes of Health (U.S.)/history , Neurosciences/history , History, 20th Century , History, 21st Century , Humans , United States
3.
Glia ; 64(7): 1190-209, 2016 07.
Article in English | MEDLINE | ID: mdl-27100937

ABSTRACT

Axonal pathology is a key contributor to long-term disability in multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS), but the mechanisms that underlie axonal pathology in MS remain elusive. Evidence suggests that axonal pathology is a direct consequence of demyelination, as we and others have shown that the node of Ranvier disassembles following loss of myelin. In contrast to the node of Ranvier, we now show that the axon initial segment (AIS), the axonal domain responsible for action potential initiation, remains intact following cuprizone-induced cortical demyelination. Instead, we find that the AIS is disrupted in the neocortex of mice that develop experimental autoimmune encephalomyelitis (EAE) independent of local demyelination. EAE-induced mice demonstrate profound compromise of AIS integrity with a progressive disruption that corresponds to EAE clinical disease severity and duration, in addition to cortical microglial reactivity. Furthermore, treatment with the drug didox results in attenuation of AIS pathology concomitantly with microglial reversion to a less reactive state. Together, our findings suggest that inflammation, but not demyelination, disrupts AIS integrity and that therapeutic intervention may protect and reverse this pathology. GLIA 2016;64:1190-1209.


Subject(s)
Axon Initial Segment/physiology , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/physiology , Microglia/metabolism , Animals , Animals, Genetically Modified , Autoimmune Diseases of the Nervous System/chemically induced , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/pathology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Death/physiology , Cells, Cultured , Cuprizone/toxicity , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation/drug effects , Hydroxamic Acids/therapeutic use , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Monoamine Oxidase Inhibitors/toxicity , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
J Biol Chem ; 288(16): 11066-73, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23457304

ABSTRACT

Neurofibromatosis type 1-derived Schwann cells isolated from malignant peripheral nerve sheath tumors (MPNSTs) overexpress PDGF receptor-ß and generate an aberrant intracellular calcium increase in response to PDGF-BB. Using the human MPNST Schwann cell line ST88-14, we demonstrate that, in addition to a transient phosphorylation of Akt, PDGF-BB stimulation produces an atypical sustained phosphorylation of Akt that is dependent on calcium and calmodulin (CaM). The sustained Akt phosphorylation did not occur in PDGF-BB-stimulated normal human Schwann cells or ST88-14 cells stimulated with stem cell factor, whose receptor is also overexpressed in ST88-14 cells. The sustained Akt phosphorylation induced by PDGF-BB was inhibited by pretreatment of the cells with either the intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl) ester (BAPTA-AM) or the CaM antagonist W7, whereas the transient portion was not inhibited. Akt also co-immunoprecipitated with CaM in a PDGF-BB-dependent manner, suggesting that direct interaction between Akt and CaM is involved in the sustained phosphorylation of Akt. Furthermore, we provide evidence that anti-apoptotic effects of PDGF-BB on serum-deprived ST88-14 cells can be inhibited by W7, implicating the PDGF-BB-induced activation of calcium/CaM in promoting cell survival, presumably through sustained Akt activation. We conclude that the activation of the calcium/CaM/Akt pathway resulting from stimulation of overexpressed PDGF receptor-ß may contribute to the survival and tumorigenicity of MPNST cells.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Calcium/metabolism , Calmodulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Schwann Cells/metabolism , Becaplermin , Calmodulin/antagonists & inhibitors , Cell Line , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Schwann Cells/pathology , Sulfonamides/pharmacology
5.
J Neuroimmunol ; 246(1-2): 58-64, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22464399

ABSTRACT

Guillain-Barré syndrome (GBS) is an autoimmune-mediated disease triggered by a preceding infection. A substantial body of evidence implicates antibodies to various gangliosides in subtypes of GBS. A significant proportion of patients with acute demyelinating subset of GBS have IgG antibodies against peripheral nervous system myelin specific neolactogangliosides such as LM1 and Hex-LM1. Although anti-neolactoganglioside antibodies in GBS were described more than two decades ago, their pathogenic role in neuropathy remains unknown due to the lack of suitable experimental models. In this study, we immunized ten guinea pigs with purified LM1 ganglioside mixed with keyhole limpet hemocyanin (KLH) and emulsified in complete Freund's adjuvant (CFA). Control guinea pigs were injected with KLH emulsified in CFA only. The animals were bled every four week intervals. The animals were boosted 3 times every four weeks. Experiments were terminated four months after initial immunization. Nine of 10 guinea pigs immunized with LM1 exhibited antibody responses to LM1. Anti-LM1 IgG titers in nine guinea pigs ranged from 1:400 to 1:12,800 at 16-weeks after initial immunization. Anti-LM1 antibodies were predominantly of IgG2 subclass. One guinea pig with the highest levels of IgG antibodies exhibited mild signs of neuropathy. There was no evidence of demyelination or inflammation in the sciatic nerves of LM1-immunized guinea pigs. Anti-LM1 antibodies bound to rat sciatic nerve myelin and to isolated rat Schwann cells. In summary, our findings suggest that relatively high levels of anti-LM1 IgG antibodies can be induced in guinea pigs and that LM1 is localized in peripheral nerve myelin and in Schwann cells. Further studies are needed to determine the pathogenic potential of anti-neolactoganglioside antibodies in neuropathy.


Subject(s)
Autoantibodies/biosynthesis , Gangliosides/immunology , Immunoglobulin G/biosynthesis , Animals , Autoantibodies/metabolism , Demyelinating Diseases/diagnosis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Gangliosides/administration & dosage , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/pathology , Guinea Pigs , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Immunization/methods , Immunoglobulin G/metabolism , Schwann Cells/immunology , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/immunology , Sciatic Nerve/pathology
6.
ASN Neuro ; 3(1): e00053, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21345173

ABSTRACT

The ability of an AEF (axolemma-enriched fraction) to influence the proliferation, survival and differentiation of OPC (oligodendrocyte progenitor cells) was evaluated. Following addition of AEF to cultured OPC, the AEF associated with the outer surface of OPC so that subsequent metabolic events were likely mediated by direct AEF-OPC contact. Addition of AEF to the cultured OPC resulted in a dose- and time-dependent increase in proliferation that was partially dependent on Akt (protein kinase B) and MAPK (mitogen-activated protein kinase) activation. The major mitogen in an AEF-SE (soluble 2.0 M NaCl extract of the AEF) was identified as aFGF (acidic fibroblast growth factor) and accounted for 50% of the mitogenicity. The remaining 50% of the mitogenicity had properties consistent with bFGF (basic fibroblast growth factor) but was not unequivocally identified. Under conditions that limit the survival of OPC in culture, AEF treatment prolonged the survival of the OPC. Antigenic and morphological examination of the AEF-treated OPC indicated that the AEF treatment helped the OPC survive in a more immature state. The potential downstream metabolic pathways potentially activated in OPC by AEF and the consequences of these activated pathways are discussed. The results of these studies are consistent with the view that direct contact of axons with OPC stimulates their proliferation and survival while preventing their differentiation.


Subject(s)
Cellular Structures/metabolism , Oligodendroglia/physiology , Schwann Cells/chemistry , Stem Cells/drug effects , Stem Cells/physiology , Animals , Animals, Newborn , Cell Count/methods , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Chromatography, Agarose/methods , Hot Temperature , Mitogen-Activated Protein Kinases , Mitogens/pharmacology , Neurons/physiology , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Stem Cells/immunology , Trypsin/pharmacology
7.
Brain Res ; 1369: 10-20, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21044615

ABSTRACT

In the developing PNS, axonal neuregulin-1 (NRG1) type III is the key determinant for myelination. However, the specific role for NRG1 (III) in the CNS has not been established. To address this issue, isotype-specific antibodies were generated, characterized, and used for the immunofluorescent localization of NRG1 (III) in the developing and adult CNS of rat. In contrast to adult peripheral nerve, which showed robust axonal staining, no immunoreactivity was observed in CNS myelinated tracts during the period of active myelination or in the adult CNS. Surprisingly, NRG1 (III) was prominently expressed on dendrites and soma in both the developing and adult CNS. These findings were corroborated through the subcellular fractionation of adult rat brain combined with an immunoblotting analysis. The immunolocalization of NRG1 (III) suggests that it plays a novel role in the myelination fate of CNS axons possibly through undetermined roles in neuronal maturation, or dendritic development and activation.


Subject(s)
Central Nervous System/metabolism , Neuregulin-1/biosynthesis , Neurogenesis/physiology , Neurons/metabolism , Peripheral Nervous System/metabolism , Animals , Antibody Specificity , Female , Fluorescent Antibody Technique , Immunoblotting , Immunoprecipitation , Male , Mice , Neuregulin-1/analysis , Protein Isoforms/analysis , Protein Isoforms/biosynthesis , Rats , Rats, Sprague-Dawley
9.
Neurochem Res ; 34(1): 17-22, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18465224

ABSTRACT

Neuregulins (NRGs) are a family of growth factors which bind to the erbB family of tyrosine kinase receptors. The exact nature and interaction of specific NRG isoforms and erbB receptors that occur during the development of the nervous system have not been reported. In order to better understand the role that different NRG isoforms and erbB receptors play in the differentiation, proliferation, and survival of neurons and glial cells, we isolated protein and mRNA from dorsal root ganglia of rat pups between embryonic day (E) 13 and postnatal day (P) 15. The relative expression levels of the NRGs and erbB receptors for the different time points were compared using both Western and RT-PCR analyses. NRG1-type1alpha protein levels were highest at E-13 and then decreased by approximately 40% and remained constant through P-15. In contrast, mRNA levels for NRG1-type1alpha remained constant from E-15 to P-15. The protein levels for NRG1-type 1beta were similar to NRG1-type1alpha at E-13 with an approximate 40% increase in the levels at E-15 and E-17 followed by a decrease to E-13 levels for the remainder of the developmental time periods. The mRNA levels for NRG1-type1beta remained constant from E-15 to P-15. The protein and mRNA expression patterns for each erbB receptor were distinctive. The protein levels for erbB-2 were highest at E-19 while erbB-3 levels were highest at E-17 and E-18. ErbB-4 protein levels were highest at E-13 and decreased through P-15. The developmental pattern for erbB-2 and erbB-4 mRNA levels had no relation to that of the corresponding protein levels while the mRNA levels for erbB-3 were highest at E-17 and E-18 similar to the pattern observed for the erbB-3 protein levels. We concluded that both NRG and erbB expression in dorsal root ganglia are mostly translationally controlled and that NRG1 isoforms and their erbB receptors are not coordinately regulated.


Subject(s)
Ganglia, Spinal/metabolism , Neuregulin-1/biosynthesis , Animals , ErbB Receptors/biosynthesis , Ganglia, Spinal/growth & development , Gene Expression Regulation, Developmental , Protein Isoforms/biosynthesis , Rats , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3/biosynthesis , Receptor, ErbB-4
10.
J Neurosci Res ; 85(6): 1347-57, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17335073

ABSTRACT

Plexiform neurofibromas are peripheral nerve sheath tumors that arise frequently in neurofibromatosis type 1 (NF1) and have a risk of malignant progression. Past efforts to establish xenograft models for neurofibroma involved the implantation of tumor fragments or heterogeneous primary cultures, which rarely achieved significant tumor growth. We report a practical and reproducible animal model of plexiform-like neurofibroma by xenograft of an immortal human NF1 tumor-derived Schwann cell line into the peripheral nerve of scid mice. The S100 and p75 positive sNF94.3 cell line was shown to possess a normal karyotype and have apparent full-length neurofibromin by Western blot. These cells were shown to have a constitutional NF1 microdeletion and elevated Ras-GTP activity, however, suggesting loss of normal neurofibromin function. Localized intraneural injection of the cell line sNF94.3 produced consistent and slow growing tumors that infiltrated and disrupted the host nerve. The xenograft tumors resembled plexiform neurofibromas with a low rate of proliferation, abundant extracellular matrix (hypocellularity), basal laminae, high vascularity, and mast cell infiltration. The histologic features of the developed tumors were particularly consistent with those of human plexiform neurofibroma as well. Intraneural xenograft of sNF94.3 cells enables the precise initiation of intraneural, plexiform-like tumors and provides a highly reproducible model for the study of plexiform neurofibroma tumorigenesis. This model facilitates testing of potential therapeutic interventions, including angiogenesis inhibitors, in a relevant cellular environment.


Subject(s)
Cell Line, Tumor , Lung Neoplasms/pathology , Neurofibromatosis 1/pathology , Schwann Cells/cytology , Adult , Animals , Blotting, Western , Female , Humans , Mice , Mice, SCID , Neoplasm Transplantation/methods , Nerve Tissue Proteins/metabolism , Neurofibromin 1/genetics , Schwann Cells/enzymology , Transplantation, Heterologous/methods , ras Proteins/metabolism
11.
J Neurosci Res ; 85(6): 1159-69, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17335081

ABSTRACT

We have shown previously that prostaglandin E(2) (PGE(2)) and prostaglandin I(2) (PGI(2)) are each produced in an explant model of peripheral nerve injury. We report that IP prostanoid receptor mRNA and protein are present in primary rat Schwann cells. IP prostanoid receptor stimulation using prostacyclin produced an elevation in intracellular cyclic AMP concentration ([cAMP](i)) in primary Schwann cells. Peak [cAMP](i) was observed between 5-15 min of stimulation followed by a gradual recovery toward basal level. Phosphorylation of cyclic AMP-response element binding protein (CREB) on Ser(133) was also detected after IP prostanoid receptor stimulation and CREB phosphorylation was inhibited completely by the protein kinase A inhibitor, H-89. Intracellular calcium levels were not affected by IP prostanoid receptor stimulation. Unlike forskolin, IP prostanoid receptor stimulation did not significantly augment Schwann cell proliferation in response to growth factor treatment. However, IP prostanoid receptor stimulation increased the number of Schwann cells that were able to generate a calcium transient in response to P2 purinergic receptor activation. These findings suggest that signaling via the IP prostanoid receptor may by relevant to Schwann cell biology in vivo.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation/physiology , Intracellular Fluid/metabolism , Receptors, Prostaglandin/metabolism , Schwann Cells/metabolism , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors , Epoprostenol/pharmacology , Gene Expression Regulation/drug effects , Intracellular Fluid/drug effects , Isoquinolines/pharmacology , Phosphorylation/drug effects , Platelet Aggregation Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, Prostaglandin/genetics , Schwann Cells/drug effects , Sciatic Nerve/cytology , Serine/metabolism , Sulfonamides/pharmacology
12.
J Immunol ; 177(8): 5226-35, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015708

ABSTRACT

CD1d-restricted NKT cells expressing invariant TCR alpha-chains (iNKT cells) produce both proinflammatory and anti-inflammatory cytokines rapidly upon activation, and are believed to play an important role in both host defense and immunoregulation. To address the potential implications of iNKT cell responses for infectious or inflammatory diseases of the nervous system, we investigated the expression of CD1d in human peripheral nerve. We found that CD1d was expressed on the surface of Schwann cells in situ and on primary or immortalized Schwann cell lines in culture. Schwann cells activated iNKT cells in a CD1d-dependent manner in the presence of alpha-galactosylceramide. Surprisingly, the cytokine production of iNKT cells stimulated by alpha-galactosylceramide presented by CD1d+ Schwann cells showed a predominance of Th2-associated cytokines such as IL-5 and IL-13 with a marked deficiency of proinflammatory Th1 cytokines such as IFN-gamma or TNF-alpha. Our findings suggest a mechanism by which iNKT cells may restrain inflammatory responses in peripheral nerves, and raise the possibility that the expression of CD1d by Schwann cells could be relevant in the pathogenesis of infectious and inflammatory diseases of the peripheral nervous system.


Subject(s)
Antigens, CD1/analysis , Cell Communication/immunology , Killer Cells, Natural/immunology , Schwann Cells/immunology , Antigens, CD1d , Cells, Cultured , Coculture Techniques , Cytokines/analysis , Galactosylceramides/pharmacology , Humans , Immunity , Inflammation , Killer Cells, Natural/cytology , Nervous System Diseases/immunology , Nervous System Diseases/pathology , Schwann Cells/chemistry , Schwann Cells/cytology , T-Lymphocytes
13.
J Neurosci Res ; 82(4): 465-71, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16235251

ABSTRACT

The growth factor receptor c-Kit has several well-characterized functions during the development of numerous cell types, including red blood cells, mast cells, and melanocytes. Its role in Schwann cells has been described in transformed cells derived from malignant peripheral nerve sheath tumors from patients with neurofibromatosis type 1 (NF1 MPNST; Badache et al. [1998] Oncogene 17:795-800). However, c-Kit functions have not been investigated in normal Schwann cells. We report here that neonatal rat Schwann cells express low c-Kit levels, whereas expression levels for c-Kit are high for Schwann cells derived from MPNST of NF1 patients. In addition, c-Kit expression is not detectable in normal adult human Schwann cells. Although the c-Kit ligand stem cell factor (SCF) induces the phosphorylation of protein kinase B (or Akt) and prevents apoptosis in Schwann cells, SCF has no effect on the proliferation or differentiation of Schwann cells.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Gene Expression/physiology , Neurofibromatoses , Proto-Oncogene Proteins c-kit/metabolism , Schwann Cells/metabolism , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Blotting, Northern , Blotting, Western/methods , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Nick-End Labeling/methods , Morpholines/pharmacology , Neuregulin-1/pharmacology , Neurofibromatoses/metabolism , Neurofibromatoses/pathology , Neurofibromin 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/pharmacology , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Schwann Cells/drug effects , Time Factors
14.
J Neurosci Res ; 79(3): 318-28, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15602756

ABSTRACT

Neurofibromatosis type 1 (NF1) is a genetic disease caused by the loss of neurofibromin, which can lead to formation of highly invasive malignant peripheral nerve sheath tumors (MPNST). We characterized platelet-derived growth factor-beta (PDGF-beta) receptor expression levels and signal transduction pathways in NF1 MPNST cell lines and compared them with the expression of PDGF-beta receptors in normal human Schwann cells (nhSC). As examined by Western blotting, PDGF-beta receptor expression levels were similar in nhSC and NF1 MPNST cell lines. MAPK and Akt also were phosphorylated in both cell types to a similar degree in response to PDGF B chains (PDGF-BB). However, increased intracellular calcium (Ca2+) levels in response to PDGF-BB were observed only in the NF1 MPNST cell lines; nhSC did not show any increase in intracellular calcium when stimulated with PDGF-BB. The calcium response in NF1 MPNST cell lines was blocked with thapsigargin, suggesting that the PDGF-BB-stimulated increases in intracellular calcium originated in the internal compartment of the cell rather than reflecting influx of calcium from the extracellular compartment. Calmodulin kinase II (CAMKII) is phosphorylated in response to PDGF-BB in the NF1 MPNST cell lines, whereas no phosphorylation of CAMKII was observed in nhSCs. The decreased growth of NF1 MPNST cell lines after treatment with a CAMKII inhibitor is consistent with the view that aberrant activation of the calcium-signaling pathway by PDGF-BB contributes to the formation of MPNST in NF1 patients.


Subject(s)
Calcium Signaling/physiology , Nerve Sheath Neoplasms/metabolism , Neurofibromatosis 1/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Schwann Cells/metabolism , Adult , Becaplermin , Calcium/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , MAP Kinase Signaling System/physiology , Male , Middle Aged , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/physiopathology , Neurofibromatosis 1/physiopathology , Neurofibromin 1/biosynthesis , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor beta/drug effects , Schwann Cells/drug effects , Schwann Cells/pathology
16.
Glia ; 46(2): 116-29, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15042580

ABSTRACT

Sciatic nerve explants cultured either alone or in the presence of peritoneal macrophages were used to study prostaglandin E(2) (PGE(2)) and 6-keto-PGF(1alpha) production following traumatic peripheral nerve injury. Although barely detectable at early time points (1-3 h in vitro), the production of PGE(2) and 6-keto-PGF(1alpha) by sciatic nerve explants increased significantly after 18 h and remained elevated for up to 96 h. The cyclooxygenase-2 (COX-2) selective inhibitor, NS-398, inhibited PGE(2) and 6-keto-PGF(1alpha) production by injured sciatic nerve in a dose-dependent manner. Consistent with the observed effect of NS-398, peripheral nerve explants, as well as Schwann cells and perineural fibroblasts cultured from neonatal sciatic nerve, each contained COX-2 immunoreactivity after 24 h in vitro. Both Schwann cells and perineural fibroblasts produced significant amounts of PGE(2) and 6-keto-PGF(1alpha); but only in the presence of arachidonic acid. As observed for injured sciatic nerve, the production of PGE(2) and 6-keto-PGF(1alpha) by primary Schwann cells and perineural fibroblasts was completely inhibited by NS-398. Compared to macrophages cultured alone, macrophages cultured in the presence of sciatic nerve explants produced large amounts of PGE(2), whereas the level of 6-keto-PGF(1alpha) was unchanged. In contrast, macrophages treated with adult sciatic nerve homogenate did not produce significant amounts of either PGE(2) or 6-keto-PGF(1alpha) during the entire course of treatment. We conclude that injured sciatic nerves produce PGE(2) and 6-keto-PGF(1alpha) by a mechanism involving COX-2 activity and that macrophages produce large amounts of PGE(2) in response to soluble factors produced by injured nerve but not during the phagocytosis of peripheral nerve debris.


Subject(s)
6-Ketoprostaglandin F1 alpha/metabolism , Dinoprostone/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2 , Fibroblasts/cytology , Fibroblasts/metabolism , In Vitro Techniques , Isoenzymes/metabolism , Macrophages/cytology , Macrophages/metabolism , Phagocytosis/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/cytology , Solubility
17.
Glia ; 45(2): 197-207, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14730713

ABSTRACT

Neuregulins (NRGs) are growth factors present in neurons and glial cells of the central and peripheral nervous systems and play a role in the survival, proliferation, and differentiation of these cells. We now report the localization of the two major isoforms of NRG (alpha and beta) and their receptors (erbB) in cultured Schwann cells and oligodendrocytes isolated from neonatal rat pups. Immunocytochemistry and Western blots for NRG and erbB receptors in defined subcellular fractions were utilized to assess cellular localization. Less differentiated oligodendrocytes contain both NRG isoforms in the cell bodies but not the processes, while only NRG-1beta was found in the nucleus. In contrast, more differentiated oligodendrocytes contained neither isoform in the nucleus while both isoforms were colocalized in the cytoplasm and cell processes. In Schwann cells, both NRG-1beta and NRG-1alpha were colocalized in the cytoplasm and processes. The Schwann cell nucleus had weak immunoreactivity for both NRG-1 isoforms, although NRG-1beta was predominant. ErbB2 and erbB3 receptors, which transduce the NRG-1 signal in Schwann cells, were found throughout the cytoplasm and in the processes and were also localized in the cell nucleus. The nuclear localization of NRG-1 isoforms and/or erbB receptors in both cell types was confirmed by Western blotting of nuclear and cytoplasmic extracts. Stimulation of Schwann cells with mitotic agents increased NRG-1beta expression in the nucleus and dramatically suppressed NRG-1alpha expression throughout the cell. The functional implications of this differential localization in myelinating cells are discussed.


Subject(s)
Myelin Sheath/metabolism , Neuregulin-1/metabolism , Neuroglia/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Animals , Animals, Newborn , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , ErbB Receptors/metabolism , Mitogens/pharmacology , Nervous System/cytology , Nervous System/growth & development , Nervous System/metabolism , Neuroglia/cytology , Neuroglia/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Protein Isoforms/metabolism , Rats , Schwann Cells/cytology , Schwann Cells/drug effects , Schwann Cells/metabolism
18.
Neurochem Res ; 29(11): 1999-2006, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15662834

ABSTRACT

Axonal loss is well correlated with functional deficits in Multiple Sclerosis (MS); however, the molecular mechanisms that underlie this axonal loss are not understood. In this review we summarize evidence that antibodies to axolemma-enriched fractions (AEF) isolated from CNS myelinated axons may play a role in axonal destruction. AEF contains potent antigens that elicit high-titer antisera, which destroy neurites in vitro, prevent neurite outgrowth, cause reactive changes in the neuronal cell bodies of origin and prevent myelination. We propose that these AEF antigens are cryptic because they are shielded from immune surveillance in vivo via the tightly sealed paranodal loops of myelin. Antibodies to AEF are found in cerebrospinal fluid (CSF) and sera of MS patients at higher levels compared with CSF or sera derived from patients with other neurological diseases. The potential identity of these cryptic antigens and their role in the axonal destruction characteristic of MS is discussed.


Subject(s)
Antigens/immunology , Axons/immunology , Axons/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Autoantibodies/immunology , Humans
19.
Empl Benefits J ; 28(3): 30-4, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12964530

ABSTRACT

One up-and-coming approach to controlling health care costs is complementary health care, which does not rely on advances in high-tech, invasive technology or expensive new pharmaceuticals, but rather focuses much more on the high-touch, direct practitioner care. It often offers lower cost alternatives to traditional medicine.


Subject(s)
Complementary Therapies/economics , Health Benefit Plans, Employee/organization & administration , Cost Control , Evidence-Based Medicine , Health Promotion , Humans , Life Style , Power, Psychological , United States
20.
Cancer Res ; 62(24): 7357-63, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12499280

ABSTRACT

Neuroblastoma (NB), a common pediatric neoplasm, consists of two main cell populations: neuroblastic/ganglionic cells and Schwann cells. NB tumors with abundant Schwannian stroma display a more benign clinical behavior than stroma-poor tumors. Recent studies suggest that Schwann cells influence NB tumor growth via secreted factors that induce differentiation, suppress proliferation, and inhibit angiogenesis. Two angiogenesis inhibitors, pigment epithelium-derived factor and tissue inhibitor of metalloproteinase-2, have been detected in Schwann cell secretions. Here, we isolated another Schwann cell-derived secreted inhibitor of angiogenesis, a 43-kDa protein identified as SPARC (secreted protein acidic and rich in cysteine), an extracellular matrix protein. We found SPARC to be critical for the antiangiogenic phenotype of cultured Schwann cells. We also show that purified SPARC potently inhibits angiogenesis and significantly impairs NB tumor growth in vivo. SPARC may be an effective candidate for the treatment of children with clinically aggressive, Schwannian stroma-poor NB tumors.


Subject(s)
Angiogenesis Inhibitors/physiology , Neovascularization, Pathologic/pathology , Neuroblastoma/blood supply , Osteonectin/physiology , Schwann Cells/metabolism , Adult , Angiogenesis Inhibitors/biosynthesis , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cell Movement/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Osteonectin/biosynthesis , Osteonectin/metabolism , Osteonectin/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...