Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 12(5): 6990-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351771

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer­related mortality among American males. Studies suggest that cigarette smoking is associated with the progression of PCa; however, the molecular mechanisms underlying this process have not been extensively investigated. PCa progression is characterized by increased cell migration and alterations in extracellular matrix (ECM)­ and cell adhesion molecule (CAM)­related gene expression. In the present study, the influence of cigarette smoke medium (SM) on cell migration and on the expression of ECM­ and CAM­related genes in PC3 prostate adenocarcinoma cells was investigated. According to a wound­healing assay, SM treatment promoted PC3 cell migration. RNA expression levels from SM­treated and control cells were analyzed using a polymerase chain reaction (PCR) array. Of 84 genes analyzed, 27.38% (23/84) exhibited a ≥2­fold change in threshold cycle in PC3 cells following 0.5% SM treatment. Functional gene grouping analysis demonstrated that SM treatment modulated the RNA transcription of approximately 18.4% of CAMs and 33.93% of ECM­related genes. Quantitative PCR analysis showed that SM treatment led to a significant decrease in transcription levels of the following genes: Collagen 5 α­1(V), connective tissue growth factor, integrin ß­2, kallmann syndrome 1, laminin α 3, matrix metallopeptidase 7 (MMP7), MMP13, secreted protein acidic cysteine­rich, thrombospondin­2 and versican; and that SM significantly increased the transcription levels of MMP2 and MMP12. Furthermore, MMP2 knockdown significantly reduced the migration of SM­treated PC3 cells. The present study provides novel insights into the association of cigarette smoking with PCa progression, via the alteration of ECM/CAM interactions.


Subject(s)
Adenocarcinoma/genetics , Cell Adhesion Molecules/genetics , Cell Movement , Extracellular Matrix/genetics , Prostate/pathology , Prostatic Neoplasms/genetics , Smoking/adverse effects , Adenocarcinoma/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/pathology
2.
Cancers (Basel) ; 6(2): 1138-56, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24830349

ABSTRACT

Tobacco use is a major public health problem worldwide. Tobacco-related cancers cause millions of deaths annually. Although several tobacco agents play a role in the development of tumors, the potent effects of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and N'-nitrosonornicotine (NNN) are unique. Metabolically activated NNK and NNN induce deleterious mutations in oncogenes and tumor suppression genes by forming DNA adducts, which could be considered as tumor initiation. Meanwhile, the binding of NNK and NNN to the nicotinic acetylcholine receptor promotes tumor growth by enhancing and deregulating cell proliferation, survival, migration, and invasion, thereby creating a microenvironment for tumor growth. These two unique aspects of NNK and NNN synergistically induce cancers in tobacco-exposed individuals. This review will discuss various types of tobacco products and tobacco-related cancers, as well as the molecular mechanisms by which nitrosamines, such as NNK and NNN, induce cancer.

3.
Int J Oncol ; 42(6): 1919-28, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23591596

ABSTRACT

Prostate cancer is the second leading cause of male-cancer related death in the United States. Despite a number of evidence-based studies which strongly suggest an association between cigarette smoking and prostate cancer, the underlying biological mechanism is largely unknown. Heme oxygenase 1 (HO-1) has been implicated in maintaining cellular homeostasis, but also in tumor angiogenesis. Nuclear HO-1 protein expression has been observed in various types of tumors including prostate cancer. These studies, however, were reported as clinical and pathological observations, and failed to investigate nuclear HO-1 at the molecular level in cancer. The present study explores the relationship between cigarette smoke and nuclear HO-1-modulated promotion of vascular endothelial growth factor (VEGF) secretion. We have demonstrated that cigarette smoke medium (SM)-induced HO-1 mRNA expression and upregulated HO-1 protein levels in the prostate cancer cell lines DU145 and PC3. We also observed that SM significantly induced nuclear expression of HO-1, and enhanced secretion of VEGF in cells. Nuclear-directed expression of HO-1 activated the transcriptional activity of VEGF and promoted VEGF secretion in prostate cancer cells. This study provides new insights into the molecular mechanism by which cigarette smoke-induced nuclear translocation of HO-1 promotes VEGF secretion in prostate cancer cells. Nuclear HO-1 may, therefore, constitute an attractive therapeutic target to inhibit angiogenesis and the progression of prostate cancer.


Subject(s)
Heme Oxygenase-1/metabolism , Prostatic Neoplasms/metabolism , Smoking/adverse effects , Vascular Endothelial Growth Factor A/metabolism , Base Sequence , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , HEK293 Cells , Heme Oxygenase-1/genetics , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Transport/drug effects , Vascular Endothelial Growth Factor A/genetics
4.
J Biol Chem ; 285(34): 26190-8, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20511222

ABSTRACT

Reactive molecules have diverse effects on cells and contribute to several pathological conditions. Cells have evolved complex protective systems to neutralize these molecules and restore redox homeostasis. Previously, we showed that association of nuclear factor (NF)-erythroid-derived 2 (E2)-related factor 2 (NRF2) with the nuclear matrix protein NRP/B was essential for the transcriptional activity of NRF2 target genes in tumor cells. The present study demonstrates the molecular mechanism by which NRP/B, via NRF2, modulates the transcriptional activity of antioxidant response element (ARE)-driven genes. NRP/B is localized in the nucleus of primary brain tissue and human neuroblastoma (SH-SY5Y) cells. Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Treatment of NRP/B or NRF2 knockdowns with H(2)O(2) induced apoptosis. Co-expression of NRF2 with members of the Kelch protein family, NRP/B, MAYVEN, or MAYVEN-related protein 2 (MRP2), revealed that the NRF2-NRP/B complex is important for the transcriptional activity of ARE-driven genes HO1 and NAD(P)H:quinine oxidoreductase 1 (NQO1). NRP/B interaction with Nrf2 was mapped to NRF2 ECH homology 4 (Neh4)/Neh5 regions of NRF2. NRP/B mutations that resulted in low binding affinity to NRF2 were unable to activate NRF2-modulated transcriptional activity of the ARE-driven genes, HO1 and NQO1. Thus, the interaction of NRP/B with the Neh4/Neh5 domains of NRF2 is indispensable for activation of NRF2-mediated ARE-driven antioxidant and detoxifying genes that confer cellular defense against oxidative stress-induced damage.


Subject(s)
Microfilament Proteins/physiology , NF-E2-Related Factor 2/metabolism , Neuropeptides/physiology , Nuclear Matrix-Associated Proteins/physiology , Nuclear Proteins/physiology , Oxidative Stress/genetics , Transcriptional Activation , Antioxidants , Brain Chemistry , Cell Line, Tumor , Gene Expression Regulation , Humans , Neuroblastoma/chemistry , Oxidation-Reduction , Response Elements
5.
PLoS One ; 4(9): e7091, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19768112

ABSTRACT

BACKGROUND: BRCA1 is a key regulatory protein participating in cell cycle checkpoint and DNA damage repair networks. BRCA1 plays important roles in protecting numerous cellular processes in response to cell damaging signals. Transforming growth factor-beta (TGF-beta) is a potent regulator of growth, apoptosis and invasiveness of tumor cells. TFG-beta activates Smad signaling via its two cell surface receptors, the TbetaRII and ALK5/TbetaRI, leading to Smad-mediated transcriptional regulation. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report an important role of BRCA1 in modulating TGF-beta signaling during oxidative stress responses. Wild-type (WT) BRCA1, but not mutated BRCA1 failed to activate TGF-beta mediated transactivation of the TGF-beta responsive reporter, p3TP-Lux. Further, WT-BRCA1, but not mutated BRCA1 increased the expression of Smad3 protein in a dose-dependent manner, while silencing of WT-BRCA1 by siRNA decreased Smad3 and Smad4 interaction induced by TGF-beta in MCF-7 breast cancer cells. BRCA1 interacted with Smad3 upon TGF-beta1 stimulation in MCF-7 cells and this interaction was mediated via the domain of 298-436aa of BRCA1 and Smad3 domain of 207-426aa. In addition, H(2)O(2) increased the colocalization and the interaction of Smad3 with WT-BRCA1. Interestingly, TGF-beta1 induced Smad3 and Smad4 interaction was increased in the presence of H(2)O(2) in cells expressing WT-BRCA1, while the TGF-beta1 induced interaction between Smad3 and Smad4 was decreased upon H(2)O(2) treatment in a dose-dependent manner in HCC1937 breast cancer cells, deficient for endogenous BRCA1. This interaction between Smad3 and Smad4 was increased in reconstituted HCC1937 cells expressing WT-BRCA1 (HCC1937/BRCA1). Further, loss of BRCA1 resulted in H(2)O(2) induced nuclear export of phosphor-Smad3 protein to the cytoplasm, resulting decreased of Smad3 and Smad4 interaction induced by TGF-beta and in significant decrease in Smad3 and Smad4 transcriptional activities. CONCLUSIONS/SIGNIFICANCE: These results strongly suggest that loss or reduction of BRCA1 alters TGF-beta growth inhibiting activity via Smad3 during oxidative stress responses.


Subject(s)
BRCA1 Protein/metabolism , Gene Expression Regulation , Oxidative Stress , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , COS Cells , Cell Proliferation , Chlorocebus aethiops , Germ-Line Mutation , Humans , Hydrogen Peroxide/metabolism , Neoplasm Invasiveness , Smad Proteins/metabolism , Transcription, Genetic
6.
J Immunol ; 181(9): 6406-16, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941231

ABSTRACT

HIV-1 infection has significant effect on the immune system as well as on the nervous system. Breakdown of the blood-brain barrier (BBB) is frequently observed in patients with HIV-associated dementia (HAD) despite lack of productive infection of human brain microvascular endothelial cells (HBMEC). Cellular products and viral proteins secreted by HIV-1 infected cells, such as the HIV-1 Gp120 envelope glycoprotein, play important roles in BBB impairment and HIV-associated dementia development. HBMEC are a major component of the BBB. Using cocultures of HBMEC and human astrocytes as a model system for human BBB as well as in vivo model, we show for the first time that cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and significantly decreased the permeability of HBMEC as well as prevented tight junction protein down-regulation of ZO-1, claudin-5, and JAM-1 in HBMEC. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that cannabinoid agonists are able to restore the integrity of HBMEC and the BBB following insults by HIV-1 Gp120. These studies may lead to better strategies for treatment modalities targeted to the BBB following HIV-1 infection of the brain based on cannabinoid pharmacotherapies.


Subject(s)
Anti-HIV Agents/pharmacology , Arachidonic Acids/pharmacology , Brain/blood supply , Brain/virology , Cannabinoid Receptor Modulators/pharmacology , Endothelium, Vascular/virology , Glycerides/pharmacology , HIV Envelope Protein gp120/antagonists & inhibitors , HIV-1/drug effects , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/prevention & control , AIDS Dementia Complex/virology , Amidohydrolases/antagonists & inhibitors , Anti-HIV Agents/therapeutic use , Arachidonic Acids/physiology , Benzamides/pharmacology , Benzamides/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/pathology , Cannabinoid Receptor Modulators/agonists , Cannabinoid Receptor Modulators/physiology , Carbamates/pharmacology , Carbamates/therapeutic use , Cell Line , Coculture Techniques , Endocannabinoids , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Glycerides/physiology , HIV Envelope Protein gp120/physiology , HIV-1/physiology , Humans , Microcirculation/drug effects , Microcirculation/physiology , Receptor, Cannabinoid, CB1/physiology
7.
Cancer Res ; 67(18): 8596-604, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875699

ABSTRACT

The transcription factor NF-E2-related factor 2 (Nrf2) translocates into the nucleus and activates phase II genes encoding detoxification enzymes and antioxidant proteins, resulting in the protection of cells from oxidative insults. However, the involvement of Nrf2-mediated oxidative stress responses in breast cancer cells is largely unknown. Notably, during our study of the Nrf2 pathway in breast cancer cells, we observed that the nuclear matrix protein NRP/B was expressed and colocalized with Nrf2 in these cells, suggesting that NRP/B is involved in Nrf2-mediated oxidative stress responses. The expression level of NRP/B was variable in different breast cancer cells and breast cancer tissues, and was found to be localized in the nucleus. NRP/B expression was increased after exposure to the oxidative stress agent, hydrogen peroxide (H(2)O(2)), particularly in the highly aggressive MDA-MB-231 breast cancer cells. Association of NRP/B with Nrf2 in vitro and in vivo was observed in MDA-MB-231 breast cancer cells, and this association was up-regulated upon exposure to H(2)O(2), but not to sodium nitroprusside, SIN-1, and DETA-NO. NRP/B also enhanced Nrf2-mediated NAD(P)H:quinine oxidoreductase 1 promoter activity. Thus, this study reveals that NRP/B enhances oxidative stress responses in breast cancer cells via the Nrf2 pathway, identifying a novel role of nuclear matrix protein(s) in oxidative stress responses.


Subject(s)
Breast Neoplasms/metabolism , Microfilament Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Animals , Breast Neoplasms/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/pharmacology , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Neuropeptides/biosynthesis , Neuropeptides/genetics , Nitric Oxide Donors/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oxidative Stress , Transfection , Up-Regulation
8.
PLoS Med ; 4(6): e186, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17550303

ABSTRACT

BACKGROUND: While vascular endothelial growth factor (VEGF) expression in breast tumors has been correlated with a poor outcome in the pathogenesis of breast cancer, the expression, localization, and function of VEGF receptors VEGFR1 (also known as FLT1) and VEGFR2 (also known as KDR or FLK1), as well as neuropilin 1 (NRP1), in breast cancer are controversial. METHODS AND FINDINGS: We investigated the expression and function of VEGF and VEGF receptors in breast cancer cells. We observed that VEGFR1 expression was abundant, VEGFR2 expression was low, and NRP1 expression was variable. MDA-MB-231 and MCF-7 breast cancer cells, transfected with antisense VEGF cDNA or with siVEGF (VEGF-targeted small interfering RNA), showed a significant reduction in VEGF expression and increased apoptosis as compared to the control cells. Additionally, specifically targeted knockdown of VEGFR1 expression by siRNA (siVEGFR1) significantly decreased the survival of breast cancer cells through down-regulation of protein kinase B (AKT) phosphorylation, while targeted knockdown of VEGFR2 or NRP1 expression had no effect on the survival of these cancer cells. Since a VEGFR1-specific ligand, placenta growth factor (PGF), did not, as expected, inhibit the breast cancer cell apoptosis induced by siVEGF, and since VEGFR1 antibody also had no effects on the survival of these cells, we examined VEGFR1 localization. VEGFR1 was predominantly expressed internally in MDA-MB-231 and MCF-7 breast cancer cells. Specifically, VEGFR1 was found to be colocalized with lamin A/C and was expressed mainly in the nuclear envelope in breast cancer cell lines and primary breast cancer tumors. Breast cancer cells treated with siVEGFR1 showed significantly decreased VEGFR1 expression levels and a lack of VEGFR1 expression in the nuclear envelope. CONCLUSIONS: This study provides, to our knowledge for the first time, evidence of a unique survival system in breast cancer cells by which VEGF can act as an internal autocrine (intracrine) survival factor through its binding to VEGFR1. These results may lead to an improved strategy for tumor therapy based on the inhibition of angiogenesis.


Subject(s)
Apoptosis/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Flow Cytometry , Humans , Immunohistochemistry , Immunoprecipitation , Lamin Type A/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Mutation , Nuclear Envelope/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/physiology
9.
J Biol Chem ; 282(16): 12319-29, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17324934

ABSTRACT

Oligodendrocytes (OLGs) are generated by progenitor cells that are committed to differentiating into myelin-forming cells of the central nervous system. Rearrangement of the cytoskeleton leading to the extension of cellular processes is essential for the myelination of axons by OLGs. Here, we have characterized a new member of the Kelch-related protein family termed MRP2 (for Mayven-related protein 2) that is specifically expressed in brain. MRP2/KLHL1 is expressed in oligodendrocyte precursors and mature OLGs, and its expression is up-regulated during OLG differentiation. MRP2/KLHL1 expression was abundant during the specific stages of oligodendrocyte development, as identified by A2B5-, O4-, and O1-specific oligodendrocyte markers. MRP2/KLHL1 was localized in the cytoplasm and along the cell processes. Moreover, a direct endogenous association of MRP2/KLHL1 with actin was observed, which was significantly increased in differentiated OLGs compared with undifferentiated OLGs. Overexpression of MRP2/KLHL1 resulted in a significant increase in the process extension of rat OLGs, whereas MRP2/KLHL1 antisense reduced the process length of primary rat OLGs. Furthermore, murine OLGs isolated from MRP2/KLHL1 transgenic mice showed a significant increase in the process extension of OLGs compared with control wild-type murine OLGs. These studies provide insights into the role of MRP2/KLHL1, through its interaction with actin, in the process elongation of OLGs.


Subject(s)
Gene Expression Regulation , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Oligodendroglia/metabolism , ATP-Binding Cassette Transporters , Actins/metabolism , Amino Acid Sequence , Animals , Mice , Mice, Transgenic , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Binding , Rats , Sequence Homology, Amino Acid
10.
J Biol Chem ; 281(52): 40450-60, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17085437

ABSTRACT

The precise role of vascular endothelial growth factor (VEGF) in regulating integrins in brain microvascular endothelial cells is unknown. Here, we analyzed VEGF effects on integrin expression and activation in human brain microvascular endothelial cells (HBMECs). Using human cDNA arrays and ribonuclease (RNase) protection assays, we observed that VEGF up-regulated the mRNA expression of alpha(6) integrin in HBMECs. VEGF significantly increased alpha(6)beta(1) integrin expression, but not alpha(6)beta(4) integrin expression in these cells. Specific down-regulation of alpha(6) integrin expression by small interfering RNA (siRNA) oligonucleotides inhibited both the capillary morphogenesis of HBMECs and their adhesion and migration. Additionally, VEGF treatment resulted in activation of alpha(6)beta(1) integrins in HBMECs. Functional blocking of alpha(6) integrin with its specific antibody inhibited the VEGF-induced adhesion and migration as well as in vivo angiogenesis, and markedly suppressed tumor angiogenesis and breast carcinoma growth in vivo. Thus, VEGF can modulate angiogenesis via increased expression and activation of alpha(6)beta(1) integrins, which may promote VEGF-driven tumor angiogenesis in vivo.


Subject(s)
Brain/blood supply , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Integrin alpha6beta1/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/physiology , Animals , Brain/cytology , Brain/metabolism , Cell Line , Cell Line, Tumor , Endothelium, Vascular/physiopathology , Female , Humans , Integrin alpha6beta1/biosynthesis , Integrin alpha6beta1/genetics , Mice , Mice, Nude , Microcirculation/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Receptors, Vascular Endothelial Growth Factor/physiology
11.
Mol Cell Biol ; 26(22): 8371-84, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16982692

ABSTRACT

The actin-based cytoskeleton is essential for the generation and maintenance of cell polarity, cellular motility, and the formation of neural cell processes. MRP2 is an actin-binding protein of the kelch-related protein family. While MRP2 has been shown to be expressed specifically in brain, its function is still unknown. Here, we report that in neuronal growth factor (NGF)-induced PC12 cells, MRP2 was expressed along the neurite processes and colocalized with Talin at the growth cones. MRP2 mRNA and protein levels were up-regulated in PC12 cells following NGF stimulation. Moreover, treatment of PC12 cells with interfering RNAs for MRP2 and glycogen synthase kinase 3beta (GSK3beta) resulted in the inhibition of neurite outgrowth. A significant decrease in MRP2 expression levels was observed following GSK3beta inhibition, which was correlated with the inhibited neurite outgrowth, while GSK3beta overexpression was found to increase MRP2 expression levels. MRP2 interacted with GSK3beta through its NH2 terminus containing the BTB domain, and these molecules colocalized along neurite processes and growth cones in differentiated PC12 cells and rat primary hippocampal neurons. Additionally, increased associations of MRP2 with GSK3beta and MRP2 with actin were observed in the NGF-treated PC12 cells. Thus, this study provides, for the first time, insights into the involvement of MRP2 in neurite outgrowth, which occurs in a GSK3beta-dependent manner.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Microfilament Proteins/physiology , Nerve Growth Factor/pharmacology , Neurites/physiology , Neurons/physiology , Actins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Microfilament Proteins/metabolism , Models, Biological , Neurites/drug effects , Neurites/enzymology , Neurons/enzymology , Neurons/metabolism , PC12 Cells , RNA, Small Interfering/physiology , Rats , Rats, Sprague-Dawley , Receptor, trkA/metabolism , Signal Transduction , Transfection
12.
Cancer Res ; 66(11): 5757-62, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740714

ABSTRACT

Amplification of the HER-2/neu (ErbB2) gene is observed in approximately 30% of human breast cancers, correlating with a poor clinical prognosis. Src kinases are also involved in the etiology of breast cancer, and their activation was suggested to be necessary for Neu-induced oncogenesis. To address whether Src activity is essential for Neu-mediated tumorigenesis, we used a physiologic inhibitor of Src kinase activity, the Csk homologous kinase (CHK), expressed as a mammary tissue-specific transgene. Our data, using a physiologic inhibitor of Src activity (CHK), showed that blocking of Neu-induced Src activity without altering Src expression levels had no significant effects on Neu-mediated mammary tumorigenesis in vivo. This contradicts the current paradigm that activation of Src kinases is essential for Neu-induced oncogenesis. This study is the first to distinguish between the kinase-dependent and kinase-independent actions of Src and shows that its kinase-dependent properties are not requisite for Neu-induced tumorigenesis.


Subject(s)
Mammary Glands, Animal/enzymology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , src-Family Kinases/metabolism , Animals , Female , Genes, erbB-2/physiology , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation , src-Family Kinases/antagonists & inhibitors
13.
J Cell Sci ; 118(Pt 23): 5537-48, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16306221

ABSTRACT

The neuronal nuclear matrix protein, NRP/B, contains a BTB domain and kelch repeats and is expressed in primary neurons but not in primary glial cells. To examine the function of NRP/B in neurons, we analyzed the structure/function of the NRP/B-BTB domain and its role in neurite outgrowth. Based on three-dimensional modeling of NRP/B, we generated an NRP/B-BTB mutant containing three mutations in the conserved amino acids D47A, H60A and R61D that was termed BTB mutant A. BTB mutant A significantly reduced the dimerization of NRP/B compared to wild-type NRP/B. The NRP/B-BTB domain was required for nuclear localization and mediated the association of NRP/B with p110RB through the TR subdomain within the B pocket of p110RB. Overexpression of wild-type NRP/B and NRP/B-BTB domain significantly induced neurite outgrowth in PC12 cells and enhanced the G0-G1 cell population by approximately 23% compared to the control cells, whereas NRP/B-BTB mutant A reduced neurite outgrowth by 70-80%, and inhibited NRP/B-p110RB association. Single cell microinjection of NRP/B-specific antibodies also blocked the neurite outgrowth of PC12 cells upon NGF stimulation. Interference of NRP/B expression by small interfering RNA (NRP/B-siRNA) inhibited neurite outgrowth and suppressed the NGF-induced outgrowth of neurites in PC12 cells. Additionally, p110RB phosphorylation at serine residue 795 was significantly reduced in PC12 cells treated with NRP/B siRNA compared to those treated with control GFP-siRNA, indicating that p110RB is a downstream target of NRP/B. Thus, the BTB domain of NRP/B regulates neurite outgrowth through its interaction with the TR subdomain within the B pocket of p110RB, and the conserved amino acids D47A, H60A and R61D within this domain of NRP/B are crucial residues for neurite extension in neuronal cells. These findings support a role for the BTB-domain of NRP/B as an important regulator of neuronal differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Microfilament Proteins/genetics , Neurites/physiology , Neuropeptides/genetics , Nuclear Proteins/genetics , Animals , Antibodies/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Line , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Gene Expression Regulation , Humans , Microfilament Proteins/metabolism , Microfilament Proteins/pharmacology , Mutation , Neurites/drug effects , Neurites/ultrastructure , Neurons/cytology , Neurons/physiology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , PC12 Cells , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Rats
14.
J Neurochem ; 92(5): 1191-203, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15715669

ABSTRACT

Rearrangement of the cytoskeleton leading to the extension of cellular processes is essential for the myelination of axons by oligodendrocytes. We observed that the actin-binding protein, Mayven, is expressed during all stages of the oligodendrocyte lineage, and that its expression is up-regulated during oligodendrocyte differentiation. Mayven is localized in the cytoplasm and along the cell processes. Mayven also binds actin, and is involved in the cytoskeletal reorganization in oligodendrocyte precursor cells (O-2A cells) that leads to process elongation. Mayven overexpression resulted in an increase in the process outgrowth of O-2A cells and in the lengths of the processes, while microinjection of Mayven-specific antibodies inhibited process extension in these cells. Furthermore, O-2A cells transduced with recombinant retroviral sense Mayven (pMIG-W-Mayven) showed an increase in the number of oligodendrocyte processes with outgrowth, while recombinant retroviral antisense Mayven (pMIG-W-Mayven-AS) blocked O-2A process extension. Interestingly, co-localization and association of Mayven with Fyn kinase were found in O-2A cells, and these interactions were increased during the outgrowth of oligodendrocyte processes. This association was mediated via the SH3 domain ligand (a.a. 1-45) of Mayven and the SH3 domain of Fyn, suggesting that Mayven may act as a linker to bind Fyn, via its N-terminus. Thus, Mayven plays a role in the dynamics of cytoskeletal rearrangement leading to the process extension of oligodendrocytes.


Subject(s)
Cell Surface Extensions/drug effects , Gene Expression Regulation, Developmental/physiology , Microfilament Proteins/pharmacology , Nerve Tissue Proteins/pharmacology , Oligodendroglia/drug effects , Prosencephalon/cytology , Actins/metabolism , Animals , Antibodies/pharmacology , Binding Sites/physiology , Blotting, Northern/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Size/drug effects , Cell Surface Extensions/physiology , Cell Surface Extensions/virology , Cells, Cultured , Drug Interactions , Embryo, Mammalian , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Microfilament Proteins/immunology , Microinjections/methods , Microscopy, Confocal/methods , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/immunology , O Antigens/metabolism , Oligodendroglia/metabolism , Oligodendroglia/virology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Rats , Recombinant Proteins/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Subcellular Fractions/metabolism , Transfection/methods , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...