Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Med ; 3(1): 14-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24402972

ABSTRACT

Dynamin 2 (Dyn2) is essential for intracellular vesicle formation and trafficking, cytokinesis, and receptor endocytosis. In this study, we investigated the implication of Dyn2 as a prognostic marker and therapeutic target for progressive prostate cancer (PCA). We evaluated Dyn2 protein expression by immunohistochemistry in two cohorts: men with localized PCA treated by retropubic radical prostatectomy (n = 226), and men with advanced/castrate-resistant PCA (CRPC) treated by transurethral resection of prostate (TURP) (n = 253). The role of Dyn2 in cell invasiveness was assessed by in vitro and in vivo experiments using androgen-responsive and refractory PCA preclinical models. Dyn2 expression was significantly increased across advanced stages of PCA compared to benign prostate tissue (P < 0.0001). In the CRPC cohort, high Dyn2 was associated with higher Gleason score (P = 0.004) and marginally with cancer-specific mortality (P = 0.052). In preclinical models, Dyn2 gene silencing significantly reduced cell migration and invasion in vitro, as well as tumor size and lymph node metastases in vivo. In isolated PCA cells, Dyn2 was found to regulate focal adhesion turnover, which is critical for cell migration; this mechanism requires full Dyn2 compared to mutants deficient in GTPase activity. In conclusion, Dyn2 overexpression is associated with neoplastic prostate epithelium and is associated with poor prognosis. Inhibition of Dyn2 prevents cell invasiveness in androgen-responsive and -refractory PCA models, supporting the potential benefit of Dyn2 to serve as a therapeutic target for advanced PCA.


Subject(s)
Carcinogenesis/genetics , Cell Movement/genetics , Dynamin II/biosynthesis , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Disease-Free Survival , Dynamin II/genetics , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Grading , Prognosis , Prostatectomy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology
2.
Int J Cardiol ; 168(3): 2109-19, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23410488

ABSTRACT

BACKGROUND: Heart failure (HF) is approaching an epidemic proportion and has become one of the leading causes of death. It imposes a great burden on the healthcare system and society. Remodeling of cardiomyocyte membranes has a profound role in the pathogenesis of HF. However, whether dynamin (DNM), a membrane-remodeling GTPase, is associated with HF remains unclear. METHODS AND RESULTS: Here, we identified that DNM2 is necessary for the maintenance of cardiac function. Endogenous DNM2 protein levels were gradually decreased in parallel with the progression of HF in different experimental animal models. Decreased DNM2 level was also observed in the end-stage failing human heart. DNM2-deficient zebrafish exhibited signs of notable cardiac apoptosis and eventually developed severe HF. Mechanistic study showed that DNM2 downregulation caused cardiomyocyte sarcoplasmic reticulum Ca(2+) overload and subsequent mitochondria-dependent apoptosis. These events were preceded by enhanced membrane translocation of the L-type Ca(2+) channel due to DNM2 deficiency-mediated membrane trafficking dysfunction. Furthermore, prevention of cardiomyocyte Ca(2+)-mishandling largely ameliorated the DNM2 deficiency-associated cardiomyocyte apoptosis and HF. CONCLUSIONS: DNM2 mediates HF by modulating Ca(2+)-dependent apoptotic death of cardiomyocyte. The finding may shed light on the new strategy of HF treatment.


Subject(s)
Apoptosis , Calcium Channels, L-Type/metabolism , Calcium/metabolism , DNA/genetics , Dynamin II/genetics , Heart Failure/genetics , Myocytes, Cardiac/ultrastructure , Animals , Blotting, Western , Disease Models, Animal , Dynamin II/biosynthesis , Heart Failure/metabolism , Heart Failure/pathology , Humans , In Situ Nick-End Labeling , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Zebrafish/embryology
3.
J Mol Med (Berl) ; 88(4): 339-50, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127478

ABSTRACT

Dynamin 2 (DNM2) mutations cause autosomal dominant centronuclear myopathy, a rare form of congenital myopathy, and intermediate and axonal forms of Charcot-Marie-Tooth disease, a peripheral neuropathy. DNM2 is a large GTPase mainly involved in membrane trafficking through its function in the formation and release of nascent vesicles from biological membranes. DNM2 participates in clathrin-dependent and clathrin-independent endocytosis and intracellular membrane trafficking (from endosomes and Golgi apparatus). Recent studies have also implicated DNM2 in exocytosis. DNM2 belongs to the machinery responsible for the formation of vesicles and regulates the cytoskeleton providing intracellular vesicle transport. In addition, DNM2 tightly interacts with and is involved in the regulation of actin and microtubule networks, independent from membrane trafficking processes. We summarize here the molecular, biochemical, and functional data on DNM2 and discuss the possible pathophysiological mechanisms via which DNM2 mutations can lead to two distinct neuromuscular disorders.


Subject(s)
Dynamin II/physiology , Myopathies, Structural, Congenital/genetics , Animals , Axons , Cell Membrane/metabolism , Charcot-Marie-Tooth Disease/metabolism , Clathrin/chemistry , Cytoskeleton/metabolism , Dynamin II/biosynthesis , Endocytosis , Endosomes/metabolism , Exocytosis , Genes, Dominant , Golgi Apparatus/metabolism , Humans , Peripheral Nervous System Diseases/pathology
4.
Cancer Res ; 67(11): 5397-407, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545621

ABSTRACT

Melanoma is the most lethal form of skin cancer. There is a lack of effective treatments for individuals with advanced disease. Many melanomas exhibit high levels of radioresistance. The direct consequence of gamma-irradiation for most melanoma cells is growth arrest at the G2-M phase of cell cycle. However, radiation-induced signaling pathways may affect numerous additional targets in cancer cells. We show in the present study that gamma-irradiation, as well as alpha-particle exposure, dramatically increases the susceptibility of melanoma cells to recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis via up-regulation of surface TRAIL-receptor 1/receptor 2 (DR4/DR5) levels and to Fas ligand-mediated apoptosis via up-regulation of surface Fas levels. Additionally, increased dynamin-2 expression after irradiation is critically important in the translocation of death receptor to the cell surface. Moreover, sodium arsenite treatment may up-regulate expression of endogenous TRAIL and induces its translocation to cell surface and further down-regulates cFLIP levels in melanoma cells. We have evaluated the effects of sequential gamma-irradiation and arsenite treatment of melanoma cells for the induction of death signaling. Such treatment results in an efficient TRAIL-mediated apoptosis via a paracrine mechanism. These data highlight the efficacy of combined modality treatment involving radiation and arsenite in clinical management of this often fatal form of skin cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenites/pharmacology , Melanoma/drug therapy , Melanoma/radiotherapy , Skin Neoplasms/drug therapy , Skin Neoplasms/radiotherapy , Sodium Compounds/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Alpha Particles , Apoptosis/drug effects , Apoptosis/radiation effects , Arsenites/administration & dosage , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Combined Modality Therapy , Cycloheximide/pharmacology , Drug Synergism , Dynamin II/biosynthesis , Dynamin II/metabolism , Fas Ligand Protein/biosynthesis , Fas Ligand Protein/metabolism , Gamma Rays , Humans , Melanoma/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Skin Neoplasms/pathology , Sodium Compounds/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/administration & dosage
5.
Cancer Res ; 65(13): 5493-7, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994918

ABSTRACT

Specific viral targeting into intrahepatic tumors remains critical for adenovirus gene therapy in liver cancer. We previously showed that ionizing radiation increases adenovirus uptake and transgene expression in cells and colon cancer xenografts. Here, we tested whether radiation induces viral uptake through virus-cell membrane interaction. We found that radiation (8 Gy) induced adenoviral gene transfer in rat hepatocytes (WB) and human colon carcinoma cells (LoVo). This induction (24.4- and 6.5-fold, respectively) and viral uptake were significantly diminished by preincubation with antibody for Dynamin 2 but not for Coxsackie adenovirus receptor or for integrin alpha(v). Radiation-induced Dynamin 2 expression was detected by immunohistochemical staining and by increased mRNA levels for Dynamin 2 in WB (1.5-fold) and LoVo (2.2-fold) cells. Specific small interference RNA (siRNA) transfection significantly inhibited Dynamin 2 expression in various tumor cell lines (LoVo, D54, and MCF-7) and abolished the radiation induction of Dynamin 2. Likewise, radiation-induced viral gene transfer in these cells (6.5-, 5.5-, and 9.0-fold, respectively) was significantly reduced in siRNA-transfected cells (2.7-, 3.7-, and 5.0-fold, respectively). Moreover, viral uptake in LoVo tumor xenografts was significantly increased in s.c. tumors (10.9-fold) when adenovirus was given i.v. at 24 hours after tumor irradiation, coincident with an elevated Dynamin 2 expression in irradiated tumors. These data suggest that ionizing radiation induces adenovirus gene transfer in cells and tumor xenografts by regulating viral uptake, potentially through interaction with cellular Dynamin 2 and thus should provide insight into improving adenovirus targeting in tumors.


Subject(s)
Adenoviridae/radiation effects , Dynamin II/physiology , Genetic Therapy/methods , Hepatocytes/virology , Neoplasms/virology , Adenoviridae/genetics , Adenoviridae/pathogenicity , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/virology , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Breast Neoplasms/virology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Colonic Neoplasms/virology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Dynamin II/antagonists & inhibitors , Dynamin II/biosynthesis , Dynamin II/genetics , Gene Transfer Techniques , Humans , Integrin alphaV/physiology , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/therapy , RNA Interference , RNA, Small Interfering/genetics , Rats , Receptors, Virus/physiology , Transfection , Xenograft Model Antitumor Assays
6.
FASEB J ; 19(12): 1692-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16049137

ABSTRACT

Vascular endothelial growth factor receptor-2 (VEGFR-2, also known as KDR) is a receptor tyrosine kinase (RTK) regulating mitogenic, chemotactic, permeability, and survival signals in vascular endothelial cells (EC) in response to its ligand, vascular permeability factor/VEGF (VPF/VEGF), arguably the most important angiogenic cytokine. However, the compartmentalization of KDR in EC and the mechanisms regulating this process have not been well defined. Here, we demonstrate that KDR is present on the plasma membrane, on endosomes, and in the perinuclear region of EC and colocalizes with early endosomal antigen (EEA1), caveolin-1, and dynamin-2, a signal transducing GTPase involved in receptor endocytosis. Furthermore, we also observed that dynamin-2 coimmunoprecipitates with KDR and is required for EC signaling/survival. Interestingly, EC overexpressing a mutant form of dynamin deficient in GTP binding (K44A) caused a selective inhibition in KDR protein level and endosomal vesicle formation and induced cell cycle arrest by inducing p21. Taken together, our findings suggest that dynamin-2 regulates KDR expression and function and hence plays an important role in VPF/VEGF mediated angiogenesis.


Subject(s)
Dynamin II/biosynthesis , Dynamin II/physiology , Endothelium, Vascular/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism , Blotting, Western , Caveolin 1/metabolism , Cell Cycle , Cell Membrane/metabolism , Cell Proliferation , Cell Separation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Endocytosis , Endosomes/metabolism , Endothelium, Vascular/cytology , Flow Cytometry , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/chemistry , Humans , Immunoprecipitation , Membrane Proteins/metabolism , Microscopy, Fluorescence , Models, Statistical , Mutation , Neovascularization, Pathologic , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL