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.
Eur J Cell Biol ; 91(9): 694-705, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22609186

ABSTRACT

Several pathways are involved in the control of endothelial cell morphology, endothelial permeability and function in order to maintain vascular homeostasis. Here we report that protein kinase N3 (PKN3) appears to play a pivotal role in maintaining endothelial cell morphology, cell-cell junctions and motility. An RNAi-based cell biological approach in cultured human endothelial cells (HUVEC) revealed that knockdown of PKN3 expression gave rise to cells with divergent cell morphology, impaired locomotion, disturbed adherens junctions (AJ) integrity and irregular actin organization. Notably, knockdown of PKN3 cells led to improper stress fiber formation and marked adhesiveness of intercellular adherens junctions when cells became stimulated with the pro-inflammatory cytokine TNF-α. Moreover, TNF-α-induced ICAM-1 expression on the cell surface was reduced in cells with suppressed PKN3 expression. Finally, loss-of-function for PKN3 appeared to affect Pyk2 phosphorylation in endothelial cells. These observations suggest that PKN3 can be considered a novel protein implicated in remodeling the actin-adherens junction, possibly by linking ICAM-1-signaling with actin/AJ dynamics. We propose that loss of PKN3 function and concomitant aberrations in actin rearrangement may attenuate pro-inflammatory activation of endothelial cells.


Subject(s)
Actins/metabolism , Adherens Junctions/metabolism , Endothelial Cells/metabolism , Protein Kinase C/deficiency , Protein Kinase C/metabolism , Cells, Cultured , Endothelial Cells/cytology , Humans , Intercellular Adhesion Molecule-1/metabolism , Signal Transduction
2.
Clin Cancer Res ; 16(22): 5469-80, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21062934

ABSTRACT

PURPOSE: Atu027, a novel RNA interference therapeutic, has been shown to inhibit lymph node metastasis in orthotopic prostate cancer mouse models. The aim of this study is to elucidate the pharmacologic activity of Atu027 in inhibiting hematogenous metastasis to the target organ lung in four different preclinical mouse models. EXPERIMENTAL DESIGN: Atu027 compared with vehicle or control small interfering RNA lipoplexes was tested in two experimental lung metastasis models (Lewis lung carcinoma, B16V) and spontaneous metastasis mouse models (MDA-MB-435, MDA-MB-231, mammary fat pad). Different dosing schedules (repeated low volume tail vein injections) were applied to obtain insight into effective Atu027 treatment. Primary tumor growth and lung metastasis were measured, and tissues were analyzed by immunohistochemistry and histology. In vitro studies in human umbilical vein endothelial cells were carried out to provide an insight into molecular changes on depletion of PKN3, in support of efficacy results. RESULTS: Intravenous administration of Atu027 prevents pulmonary metastasis. In particular, formation of spontaneous lung metastasis was significantly inhibited in animals with large tumor grafts as well as in mice with resected primary mammary fat pad tumors. In addition, we provide evidence that an increase in VE-cadherin protein levels as a downstream result of PKN3 target gene inhibition may change endothelial function, resulting in reduced colonization and micrometastasis formation. CONCLUSION: Atu027 can be considered as a potent drug for preventing lung metastasis formation, which might be suitable for preventing hematogenous metastasis in addition to standard cancer therapy.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Carcinoma, Lewis Lung/secondary , Disease Models, Animal , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Humans , Injections, Intravenous , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Xenograft Model Antitumor Assays
3.
Exp Cell Res ; 315(13): 2165-80, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19409380

ABSTRACT

RNAi mediated loss of Drp1 function changes mitochondrial morphology in cultured HeLa and HUVEC cells by shifting the balance of mitochondrial fission and fusion towards unopposed fusion. Over time, inhibition of Drp1 expression results in the formation of a highly branched mitochondrial network along with "bulge"-like structures. These changes in mitochondrial morphology are accompanied by a reduction in levels of Mitofusin 1 (Mfn1) and 2 (Mfn2) and a modified proteolytic processing of OPA1 isoforms, resulting in the inhibition of cell proliferation. In addition, our data imply that bulge formation is driven by Mfn1 action along with particular proteolytic short-OPA1 (s-OPA1) variants: Loss of Mfn2 in the absence of Drp1 results in an increase of Mfn1 levels along with processed s-OPA1-isoforms, thereby enhancing continuous "fusion" and bulge formation. Moreover, bulge formation might reflect s-OPA1 mitochondrial membrane remodeling activity, resulting in the compartmentalization of cytochrome c deposits. The proteins Yme1L and PHB2 appeared not associated with the observed enhanced OPA1 proteolysis upon RNAi of Drp1, suggesting the existence of other OPA1 processing controlling proteins. Taken together, Drp1 appears to affect the activity of the mitochondrial fusion machinery by unbalancing the protein levels of mitofusins and OPA1.


Subject(s)
GTP Phosphohydrolases/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes , Mitochondrial Proteins/metabolism , Animals , Dynamins , GTP Phosphohydrolases/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Fusion/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/genetics , Prohibitins , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
4.
Cancer Res ; 68(23): 9788-98, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047158

ABSTRACT

We have previously described a small interfering RNA (siRNA) delivery system (AtuPLEX) for RNA interference (RNAi) in the vasculature of mice. Here we report preclinical data for Atu027, a siRNA-lipoplex directed against protein kinase N3 (PKN3), currently under development for the treatment of advanced solid cancer. In vitro studies revealed that Atu027-mediated inhibition of PKN3 function in primary endothelial cells impaired tube formation on extracellular matrix and cell migration, but is not essential for proliferation. Systemic administration of Atu027 by repeated bolus injections or infusions in mice, rats, and nonhuman primates results in specific, RNAi-mediated silencing of PKN3 expression. We show the efficacy of Atu027 in orthotopic mouse models for prostate and pancreatic cancers with significant inhibition of tumor growth and lymph node metastasis formation. The tumor vasculature of Atu027-treated animals showed a specific reduction in lymph vessel density but no significant changes in microvascular density.


Subject(s)
Pancreatic Neoplasms/therapy , Prostatic Neoplasms/therapy , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Animals , Cell Growth Processes/physiology , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/enzymology , HeLa Cells , Humans , Liposomes/administration & dosage , Lymphatic Metastasis , Macaca fascicularis , Male , Mice , Mice, SCID , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Rats , Transfection/methods
5.
Microvasc Res ; 76(1): 31-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18455200

ABSTRACT

Liposomally formulated siRNA can be used for RNAi applications in vivo. Intravenous bolus administration of lipoplexed siRNA has been shown to reduce gene expression in the vascular endothelium. Here, we applied immunofluorescence staining for different endothelial markers (PECAM-1, CD34, laminin) on paraffin sections to compare the respective expression pattern with the intracellular localization of intravenously administered, fluorescently labeled siRNA (siRNA-Cy3-lipoplex). By confocal microscopy, lipoplexed siRNA-Cy3 was detected inside vascular endothelial cells in vivo, which where identified with co-staining of endothelial markers. Consequently, the finding of intracellular siRNA uptake by vascular endothelial cells correlated with RNAi based specific protein reduction in situ as revealed by PECAM-1 specific immunofluorescence staining in lung tissue sections. Therefore, by using a cell biological approach these in situ data emphasize the functional uptake of liposomal siRNA molecules in vascular endothelial cells of different mouse tissues as indicated in our previous molecular study.


Subject(s)
Endothelial Cells/metabolism , Intracellular Space/metabolism , RNA, Small Interfering/metabolism , Animals , Antigens, CD/genetics , Antigens, CD34/analysis , Cadherins/genetics , Carbocyanines/chemistry , Endothelial Cells/chemistry , Endothelium, Lymphatic/chemistry , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Fluorescent Antibody Technique , Gene Expression , Glycoproteins/analysis , Immunohistochemistry , Intracellular Space/chemistry , Laminin/analysis , Liposomes , Liver/chemistry , Liver/cytology , Liver/metabolism , Lung/chemistry , Lung/cytology , Lung/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Nude , Myocardium/chemistry , Myocardium/cytology , Myocardium/metabolism , PTEN Phosphohydrolase/genetics , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
EMBO J ; 23(16): 3303-13, 2004 Aug 18.
Article in English | MEDLINE | ID: mdl-15282551

ABSTRACT

Chronic activation of the phosphoinositide 3-kinase (PI3K)/PTEN signal transduction pathway contributes to metastatic cell growth, but up to now effectors mediating this response are poorly defined. By simulating chronic activation of PI3K signaling experimentally, combined with three-dimensional (3D) culture conditions and gene expression profiling, we aimed to identify novel effectors that contribute to malignant cell growth. Using this approach we identified and validated PKN3, a barely characterized protein kinase C-related molecule, as a novel effector mediating malignant cell growth downstream of activated PI3K. PKN3 is required for invasive prostate cell growth as assessed by 3D cell culture assays and in an orthotopic mouse tumor model by inducible expression of short hairpin RNA (shRNA). We demonstrate that PKN3 is regulated by PI3K at both the expression level and the catalytic activity level. Therefore, PKN3 might represent a preferred target for therapeutic intervention in cancers that lack tumor suppressor PTEN function or depend on chronic activation of PI3K.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Basement Membrane/enzymology , Basement Membrane/metabolism , Basement Membrane/pathology , Catalysis , Cell Division , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms/genetics , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases/deficiency , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...