Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Exp Neurol ; 325: 113120, 2020 03.
Article in English | MEDLINE | ID: mdl-31751571

ABSTRACT

Multiple system atrophy (MSA) is a fatal disorder with no effective treatment. MSA pathology is characterized by α-synuclein (aSyn) accumulation in oligodendrocytes, the myelinating glial cells of the central nervous system (CNS). aSyn accumulation in oligodendrocytes forms the pathognomonic glial cytoplasmic inclusions (GCIs) of MSA. MSA aSyn pathology is also associated with motor and autonomic dysfunction, including an impaired ability to sweat. MSA patients have abnormal CNS expression of glial-cell-line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Our prior studies using the parent compound FTY720, a food and drug administration (FDA) approved immunosuppressive for multiple sclerosis, reveal that FTY720 protects parkinsonian mice by increasing BDNF. Our FTY720-derivative, FTY720-Mitoxy, is known to increase expression of oligodendrocyte BDNF, GDNF, and nerve growth factor (NGF) but does not reduce levels of circulating lymphocytes as it is not phosphorylated so cannot modulate sphingosine 1 phosphate receptors (S1PRs). To preclinically assess FTY720-Mitoxy for MSA, we used mice expressing human aSyn in oligodendrocytes under a 2,' 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter. CNP-aSyn transgenic (Tg) mice develop motor dysfunction between 7 and 9 mo, and progressive GCI pathology. Using liquid chromatography-mass spectrometry (LC-MS/MS) and enzymatic assays, we confirmed that FTY720-Mitoxy was stable and active. Vehicle or FTY720-Mitoxy (1.1 mg/kg/day) was delivered to wild type (WT) or Tg littermates from 8.5-11.5 mo by osmotic pump. We behaviorally assessed their movement by rotarod and sweat production by starch­iodine test. Postmortem tissues were evaluated by qPCR for BDNF, GDNF, NGF and GDNF-receptor RET mRNA and for aSyn, BDNF, GDNF, and Iba1 protein by immunoblot. MicroRNAs (miRNAs) were also assessed by qPCR. FTY720-Mitoxy normalized movement, sweat function and soleus muscle mass in 11.5 mo Tg MSA mice. FTY720-Mitoxy also increased levels of brain GDNF and reduced brain miR-96-5p, a miRNA that acts to decrease GDNF expression. Moreover, FTY720-Mitoxy blocked aSyn pathology measured by sequential protein extraction and immunoblot, and microglial activation assessed by immunohistochemistry and immunoblot. In the 3-nitropropionic acid (3NP) toxin model of MSA, FTY720-Mitoxy protected movement and mitochondria in WT and CNP-aSyn Tg littermates. Our data confirm potent in vivo protection by FTY720-Mitoxy, supporting its further evaluation as a potential therapy for MSA and related synucleinopathies.


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Multiple System Atrophy/pathology , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Female , Fingolimod Hydrochloride/pharmacology , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Transgenic , MicroRNAs/drug effects , MicroRNAs/metabolism , Multiple System Atrophy/metabolism , Proto-Oncogene Proteins c-ret/biosynthesis , Proto-Oncogene Proteins c-ret/drug effects , alpha-Synuclein/genetics
2.
Medicine (Baltimore) ; 98(3): e14120, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30653139

ABSTRACT

RATIONALE: Lung cancer is a series of gene-driven disease. EGFR, ALK, and ROS1 are 3 major driver genes that play an important role in lung cancer development and precision management. Additionally, rare genetic alterations continue to be discovered and may become novel targets for therapy. The RET gene is one of such rare genetic alteration of non-small cell lung cancer (NSCLC). In this report, we present a RET-positive case that benefited from cabozantinib treatment. PATIENT CONCERN: A 50-year-old male patient was diagnosed with lung adenocarcinoma 2 years ago, at that time he received palliative surgery of pulmonary carcinoma and completed 4 cycles of chemotherapy with gemcitabine and cisplatin. Six months later, he was hospitalized in our cancer center due to the disease recurrence, presenting with pleural metastasis. DIAGNOSIS: Gene alteration was examined using the intraoperative specimen by PCR method, and KIF5B/RET gene fusion was detected. Therefore, the patient was diagnosed with late-stage lung adenocarcinoma with RET gene mutation. INTERVENTIONS: The patient received treatment with cabozantinib from June 2017. OUTCOMES: Cabozantinib was administered (140 mg orally, once daily) for approximate 9 months, and his disease achieved stable disease (SD). During that period, there were no severe adverse events (AE), except for a grade II rash (CTCAE 4.0). LESSONS: We found that the RET fusion gene is a novel driver molecular of lung adenocarcinoma in patients without common mutations in such genes as EGFR, ALK, and ROS1. This case report supports a rationale for the treatment of lung adenocarcinoma patients with a RET fusion and provides alternative treatment options for these types of NSCLC patients.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Anilides/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/genetics , Pyridines/therapeutic use , Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Fusion/drug effects , Humans , Kinesins/genetics , Lung Neoplasms/genetics , Male , Middle Aged , Proto-Oncogene Proteins c-ret/drug effects
3.
Postgrad Med ; 126(7): 139-48, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25387222

ABSTRACT

Non-small-cell lung cancer (NSCLC) is a growing concern worldwide, and its incidence continues to increase in developing countries. It has a strong association with smoking. Lung cancer remains the leading cause of cancer-related deaths in most industrialized countries and in the United States. In the last 10 years, there have been significant advancements in the understanding of molecular oncogenes and how they play a role in driving lung cancer to both grow and metastasize. Understanding this rapidly expanding field has the potential to extend life, and it is an important field for all providers to conceptualize if they are treating patients with lung cancer. Currently, > 50% of all NSCLC is linked to 1 of several known genetic driver mutations. Using online databases, expert opinion, and practice-changing trials, we review the current standards of molecular testing of NSCLC and the expanding evidence of oncogenic drivers in nonsquamous NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Proto-Oncogenes/drug effects , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-ret/drug effects , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins p21(ras) , Proto-Oncogenes/genetics , Tomography, X-Ray Computed , ras Proteins/drug effects , ras Proteins/genetics
4.
J Neurochem ; 130(4): 541-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821093

ABSTRACT

It has been proposed that GM1 ganglioside promotes neuronal growth, phenotypic expression, and survival by modulating tyrosine kinase receptors for neurotrophic factors. Our studies tested the hypothesis that GM1 exerts its neurotrophic action on dopaminergic neurons, in part, by interacting with the GDNF (glia cell-derived neurotrophic factor) receptor complex, Ret tyrosine kinase and GFRα1 co-receptor. GM1 addition to striatal slices in situ increased Ret activity in a concentration- and time-dependent manner. GM1-induced Ret activation required the whole GM1 molecule and was inhibited by the kinase inhibitors PP2 and PP1. Ret activation was followed by Tyr1062 phosphorylation and PI3 kinase/Akt recruitment. The Src kinase was associated with Ret and GM1 enhanced its phosphorylation. GM1 responses required the presence of GFRα1, and there was a GM1 concentration-dependent increase in the binding of endogenous GDNF which paralleled that of Ret activation. Neutralization of the released GDNF did not influence the Ret response to GM1, and GM1 had no effect on GDNF release. Our in situ studies suggest that GM1 via GFRα1 modulates Ret activation and phosphorylation in the striatum and provide a putative mechanism for its effects on dopaminergic neurons. Indeed, chronic GM1 treatment enhanced Ret activity and phosphorylation in the striatum of the MPTP-mouse and kinase activation was associated with recovery of dopamine and DOPAC deficits. It has been proposed that the ganglioside GM1 promotes neuronal growth, phenotypic expression, and survival by modulating tyrosine kinase receptors for neurotrophic factors. We provide evidence that the GM1 enhances the activity of Ret tyrosine kinase receptor for glia cell-derived neurotrophic factor (GDNF) in the striatum in situ and in vivo, and propose that this might be a mechanism for GM1's neurotrophic actions on dopaminergic neurons. Ret activation is followed by Tyr1062 and Tyr981 phosphorylation and recruitment of PI3-K/Akt, Erk, and Src signaling. GM1 apparently acts by increasing the binding of endogenous GDNF to GFRα1 co-receptor, which is required for the GM1 effect on Ret.


Subject(s)
G(M1) Ganglioside/pharmacology , Neostriatum/physiology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/drug effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Blotting, Western , Dopamine/metabolism , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Immunoprecipitation , In Vitro Techniques , Male , Mice , Neostriatum/drug effects , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-ret/drug effects , src-Family Kinases/metabolism
5.
Eur J Cancer ; 50(9): 1657-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24703573

ABSTRACT

BACKGROUND: Extraskeletal myxoid chondrosarcoma (EMC) is a rare soft tissue sarcoma, marked by NR4A3 rearrangement. Herein we report on the activity of sunitinib in a series of 10 patients, strengthening what initially observed in two cases. PATIENTS AND METHODS: From July 2011, 10 patients with progressive metastatic translocated EMC have been consecutively treated with sunitinib 37.5mg/day, on a named-use basis. In an attempt to interpret the activity of sunitinib in EMC, genotype/phenotype correlations were carried out by fluorescence in situ hybridization (FISH) analyses. Moreover, transcriptome, immunohistochemical and biochemical analyses of a limited set of samples were performed focusing on some putative targets of sunitinib. RESULTS: Eight of 10 patients are still on therapy. Six patients had a Response Evaluation Criteria in Solid Tumours (RECIST) partial response (PR), two were stable, two progressed. Positron emission tomography (PET) was consistent in 6/6 evaluable cases. One patient underwent surgery after sunitinib, with evidence of a pathologic response. At a median follow-up of 8.5 months (range 2-28), no secondary resistance was detected. Median progression free survival (PFS) has not been reached. Interestingly, all responsive cases turned out to express the typical EWSR1-NR4A3 fusion, while refractory cases carried the alternative TAF15-NR4A3 fusion. Among putative sunitinib targets, only RET was expressed and activated in analysed samples. CONCLUSIONS: This report confirms the therapeutic activity of sunitinib in EMC. Genotype/phenotype analyses support a correlation between response and EWSR1-NR4A3 fusion. Involvement of RET deserves further investigation.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Bone Neoplasms , Chondrosarcoma/drug therapy , Indoles/therapeutic use , Neoplasms, Connective and Soft Tissue/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Calmodulin-Binding Proteins/genetics , Chondrosarcoma/genetics , Chondrosarcoma/secondary , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Rearrangement/genetics , Genotype , Humans , Male , Middle Aged , Neoplasms, Connective and Soft Tissue/genetics , Neoplasms, Connective and Soft Tissue/secondary , Phenotype , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-ret/drug effects , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/drug effects , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Sunitinib , TATA-Binding Protein Associated Factors/genetics , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/drug effects , fms-Like Tyrosine Kinase 3/drug effects
6.
Thyroid ; 24(2): 327-38, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23885719

ABSTRACT

BACKGROUND: RET/PTC1 is the most prevalent type of gene rearrangement found in papillary thyroid carcinoma (PTC). Previously, we introduced a new noncationic nanosystem for targeted RET/PTC1 silencing by efficient delivery of small interfering RNA (siRNA) using the "squalenoylation" approach. With the aim of improving these results further, we designed new squalenoyl nanostructures consisting of the fusogenic peptide GALA-cholesterol (GALA-Chol) and squalene (SQ) nanoparticles (NPs) of siRNA RET/PTC1. METHODS: The siRNA RET/PTC1-SQ bioconjugate was synthesized. The corresponding NPs were prepared with or without GALA-Chol by nanoprecipitation and then characterized for their size and zeta potential. The effects of NPs on BHP 10-3 SCmice and TPC-1 cell viability (MTT assay), gene and protein silencing (reverse transcription-quantitative polymerase chain reaction [rt-qPCR], Western blot), and cellular uptake (fluorescent microscopy) were studied. In vivo gene silencing efficiency of siRNA RET/PTC1-SQ NPs was assessed by administration in nude mice via either intratumoral (i.t.) or intravenous (i.v.) routes. Tumor growth was followed for 19 days. Tumors were then collected, and RET/PTC1 gene and protein inhibitions were assessed by RT-qPCR and Western blot. RESULTS: The combination of siRNA RET/PTC1-SQ bioconjugate and GALA-Chol leads to stable NPs of ∼200 nm diameter. In vitro, the results revealed that combining GALA-Chol with siRNA RET/PTC1-SQ NPs decreased cell viability, enhanced cellular internalization, and induced gene silencing efficiency in both human PTC (BHP 10-3 SCmice and TPC-1) cell lines. On the contrary, in vivo, the siRNA RET/PTC1-SQ GALA-Chol NPs were not found to be efficient either in gene silencing or in tumor growth inhibition, compared to siRNA RET/PTC1-SQ NPs both via i.t. and i.v. routes (p<0.001). CONCLUSIONS: Conversely to siRNA RET/PTC1-SQ NPs, the siRNA RET/PTC1-SQ GALA-Chol NPs are efficient in vitro but not in vivo. Finally, NPs of siRNA RET/PTC1-SQ were found to be efficient silencers of the RET/PTC1 fusion oncogene in in vivo applications even at a concentration lower than used in a previously published study.


Subject(s)
Carcinoma, Papillary/drug therapy , Carcinoma/drug therapy , Cholesterol/analogs & derivatives , Nanoparticles/administration & dosage , Peptides/administration & dosage , Proto-Oncogene Proteins c-ret/drug effects , RNA, Small Interfering/administration & dosage , Squalene/administration & dosage , Thyroid Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/administration & dosage , Gene Rearrangement , Gene Silencing , Humans , Mice , Mice, Nude , Oncogene Proteins, Fusion/drug effects , Proto-Oncogene Proteins c-ret/genetics , Thyroid Cancer, Papillary
7.
Am J Respir Crit Care Med ; 188(7): 770-5, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23841470

ABSTRACT

The remarkable success of epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors in patients with EGFR mutations and ALK rearrangements, respectively, introduced the era of targeted therapy in advanced non-small cell lung cancer (NSCLC), shifting treatment from platinum-based combination chemotherapy to molecularly tailored therapy. Recent genomic studies in lung adenocarcinoma identified other potential therapeutic targets, including ROS1 rearrangements, RET fusions, MET amplification, and activating mutations in BRAF, HER2, and KRAS in frequencies exceeding 1%. Lung cancers that harbor these genomic changes can potentially be targeted with agents approved for other indications or under clinical development. The need to generate increasing amounts of genomic information should prompt health-care providers to be mindful of the amounts of tissue needed for these assays when planning diagnostic procedures. In this review, we summarize oncogenic drivers in NSCLC that can be currently detected, highlight their potential therapeutic implications, and discuss practical considerations for successful application of tumor genotyping in clinical decision making.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma of Lung , Anaplastic Lymphoma Kinase , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/therapeutic use , Genomics , Humans , Lung Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Mutation/drug effects , Mutation/genetics , Oncogene Proteins/drug effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/drug effects , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-ret/drug effects , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins p21(ras) , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/therapeutic use , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/genetics , ras Proteins/drug effects , ras Proteins/genetics
8.
Neurosci Lett ; 417(2): 176-80, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17403572

ABSTRACT

Gial derived neurotrophic factor (GDNF) modulates neuronal cell differentiation during development and protects against neurodegeneration by preventing apoptosis at maturity. GDNF's role in tissue maintenance has generated interest in the therapeutic potential of GDNF in treating neurological disorders such as Parkinson's disease. Heparan sulfate has been shown to be essential for GDNF signaling and altering the levels of heparan sulfate promotes or inhibits GDNF functional activity. To search for other oligosaccharides capable of modulating GDNF activity as potential therapeutic molecules, we investigated the effect of acidic oligosaccharide sugar chain (AOSC) and its sulfated derivative on GDNF induced neurotrophic events by using Western-blotting, immunofluorescence cell staining, and immunoprecipitation techniques in PC12 cells expressing the GDNF receptors GFR alpha 1-Ret. AOSC significantly improved the neurite outgrowth and activated c-Ret phosphorylation in PC12-GFR alpha 1-Ret cells, but its sulfated derivative inhibited GDNF activity. Studies to understand the opposing biological effects of AOSC and its sulfated derivative on GDNF activity demonstrated that reduced GDNF binding to PC12-GFR alpha 1-Ret cell surface in the presence of the sulfated derivative likely suppressed GDNF activity as both AOSC and its sulfated derivatives had similar binding affinities to GDNF. This study illustrates the importance of oligosaccharide structure and charge on influencing GDNF activity and the potential use of oligosaccharides in modulating GDNF activity for therapeutic purposes.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/drug effects , Mannans/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Mannans/chemistry , Mannans/therapeutic use , Molecular Structure , Neurites/drug effects , Neurites/metabolism , Neurites/ultrastructure , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , PC12 Cells , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/physiology , Proto-Oncogene Proteins c-ret/drug effects , Proto-Oncogene Proteins c-ret/metabolism , Rats , Signal Transduction/physiology , Sulfuric Acid Esters/metabolism
9.
World J Surg ; 31(5): 946-56, 2007 May.
Article in English | MEDLINE | ID: mdl-17426901

ABSTRACT

Medullary thyroid carcinoma (MTC) is a rare malignancy of the thyroid C cells. It occurs in hereditary (25% of cases) and sporadic (75%) forms. Sporadic MTCs frequently metastasize to cervical lymph nodes. Thorough surgical extirpation of the primary tumor and nodal metastases by compartment-oriented resection has been the mainstay of treatment (level IV evidence). Surgical resection of residual and recurrent disease is effective in reducing calcitonin levels and controlling complications of central neck disease (level IV evidence). Radioactive iodine, external beam radiation therapy, and conventional chemotherapy have not been effective. Newer systemic treatments, with agents that target abnormal RET proteins hold promise and are being tested in clinical trials for patients with metastatic disease.


Subject(s)
Carcinoma, Medullary/therapy , Evidence-Based Medicine , Thyroid Neoplasms/therapy , Carcinoma, Medullary/pathology , Combined Modality Therapy , Diagnostic Imaging , Humans , Lymphatic Metastasis , Neck Dissection , Neoplasm Recurrence, Local , Neoplasm, Residual , Proto-Oncogene Proteins c-ret/drug effects , Thyroid Neoplasms/pathology , Thyroidectomy
10.
Nat Clin Pract Oncol ; 3(10): 564-74, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17019434

ABSTRACT

Growing evidence supports the concept of oncogene dependence for cancer development; inhibition of the initiating oncogene can result in revertion of the neoplastic phenotype. The outstanding role of the RET proto-oncogene in the development of medullary thyroid carcinoma (MTC) is well established. With the emerging knowledge concerning the signal transduction pathways leading to subsequent neoplastic transformation, oncogenic activated RET becomes a highly attractive target for selective cancer therapy. A variety of novel approaches that target RET directly or indirectly have recently emerged and an increasing number are currently being assessed in clinical trials. In view of these findings, it becomes strikingly obvious that inhibition of RET oncogene function can be a viable option for the treatment of MTC. We summarize the current evidence for RET involvement in the etiology of MTC, and the therapeutic targeting of this process in preclinical and clinical studies.


Subject(s)
Carcinoma, Medullary/genetics , Proto-Oncogene Proteins c-ret/drug effects , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/genetics , Carcinoma, Medullary/physiopathology , Carcinoma, Medullary/therapy , Cell Transformation, Neoplastic/genetics , Clinical Trials as Topic , Drug Therapy/trends , Humans , Oncogenes , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction , Thyroid Neoplasms/physiopathology , Thyroid Neoplasms/therapy
11.
Glia ; 54(6): 536-44, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16906542

ABSTRACT

Olfactory ensheathing cells (OECs) are a unique type of macroglia with axonal growth-promoting properties. The migrating ability of OECs in CNS is essential for neural regeneration. However, little is known about the extracellular and intracellular factors that regulate OEC migration. In the present study, we examined the effects of glial cell line-derived neurotrophic factor (GDNF) on OECs migration. Initially, the "scratch" migration assay, Boyden chamber assay, and explant migration assay showed that GDNF could promote OECs migration in vitro. Treatment of OECs with GDNF also induced cytoskeleton reorganization and up-regulated expression of cytoskeleton proteins. GDNF-induced OECs migration was demonstrated depending on GFRalpha-1 and Ret receptor, and activation of JNK and Src signaling cascades. Furthermore, GDNF was found to promote implanted OECs migration in a spinal cord hemisection injury model. Together, we report, to our knowledge for the first time, that GDNF stimulate OECs migration in vitro and in vivo.


Subject(s)
Cell Movement/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Olfactory Bulb/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroglia/cytology , Olfactory Bulb/cytology , Proto-Oncogene Proteins c-ret/drug effects , Proto-Oncogene Proteins c-ret/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , src-Family Kinases/drug effects , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL