Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 4: e173, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26501855

ABSTRACT

TIFA (TNF receptor associated factor (TRAF)-interacting protein with a Forkhead-associated (FHA) domain), also called T2BP, was first identified using a yeast two-hybrid screening. TIFA contains a FHA domain, which directly binds phosphothreonine and phosphoserine, and a consensus TRAF6-binding motif. TIFA-mediated oligomerization and poly-ubiquitinylation of TRAF6 mediates signaling downstream of the Tumor necrosis factor alpha receptor 1 (TNFaR-I) and interleukin-1/Toll-like receptor 4 (TLR4) pathways. Examining TIFA expression in hepatocellular carcinoma (HCC) tissues microarrays, we noted marked decreases TIFA reactivity in tumor versus control samples. In agreement, we found that HCC cell lines show reduced TIFA expression levels versus normal liver controls. Reconstituting TIFA expression in HCC cell lines promoted two independent apoptosis signaling pathways: the induction of p53 and cell cycle arrest, and the activation of caspase-8 and caspase-3. In contrast, the expression of a non-oligomerizing mutant of TIFA impacted cells minimally, and suppression of TIFA expression protected cells from apoptosis. Mice bearing TIFA overexpression hepatocellular xenografts develop smaller tumors versus TIFA mutant tumors; terminal deoxynucleotidyl transferase dUTP nick end labeling staining demonstrates increased cell apoptosis, and decreased proliferation, reflecting cell cycle arrest. Interestingly, p53 has a greater role in decreased proliferation than cell death, as it appeared dispensable for TIFA-induced cell killing. The findings demonstrate a novel suppressive role of TIFA in HCC progression via promotion of cell death independent of p53.

2.
Cell Death Dis ; 5: e1496, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25356865

ABSTRACT

Ovarian cancer patients are typically treated with carboplatin and paclitaxel, but suffer a high rate of relapse with recalcitrant disease. This challenge has fostered the development of novel approaches to treatment, including antagonists of the 'inhibitor of apoptosis proteins' (IAPs), also called SMAC mimetics, as apoptosis-inducing agents whose action is opposed by caspase inhibitors. Surprisingly, IAP antagonist plus caspase inhibitor (IZ) treatment selectively induced a tumor necrosis factor-α (TNFα)-dependent death among several apoptosis-resistant cell lines and patient xenografts. The induction of necroptosis was common in ovarian cancer, with expression of catalytically active receptor-interacting protein kinase-3 (RIPK3) necessary for death, and in fact sufficient to compromise survival of RIPK3-negative, necroptosis-resistant ovarian cancer cells. The formation of a necrosome-like complex with a second critical effector, receptor-interacting serine-threonine kinase-1 (RIPK1), was observed. RIPK1, RIPK3 and TNFα were required for the induction of death, as agents that inhibit the function of any of these targets prevented cell death. Abundant RIPK3 transcript is common in serous ovarian cancers, suggesting that further evaluation and targeting of this RIPK3-dependent pathway may be of clinical benefit.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Autocrine Communication/drug effects , Cisplatin/therapeutic use , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mutation/genetics , Necrosis , Oligopeptides/pharmacology , Ovarian Neoplasms/enzymology , Phenotype , Protease Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
3.
Curr Mol Med ; 14(2): 246-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24467204

ABSTRACT

The caspases are a family of ubiquitously expressed cysteine proteases best known for their roles in programmed cell death. However, caspases play a number of other roles in vertebrates. In the case of caspase-8, loss of expression is an embryonic lethal phenotype, and caspase-8 plays roles in suppressing cellular necrosis, promoting differentiation and immune signaling, regulating autophagy, and promoting cellular migration. Apoptosis and migration require localization of caspase-8 in the periphery of the cells, where caspase-8 acts as part of distinct biosensory complexes that either promote migration in appropriate cellular microenvironments, or cell death in inappropriate settings. In the cellular periphery, caspase-8 interacts with components of the focal adhesion complex in a tyrosine-kinase dependent manner, promoting both cell migration in vitro and metastasis in vivo. Mechanistically, caspase-8 interacts with components of both focal adhesions and early endosomes, enhancing focal adhesion turnover and promoting rapid integrin recycling to the cell surface. Clinically, this suggests that the expression of caspase-8 may not always be a positive prognostic sign, and that the role of caspase-8 in cancer progression is likely context-dependent.


Subject(s)
Caspase 8/metabolism , Cell Movement/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Caspase 8/genetics , Cell Movement/genetics , Humans
4.
Oncogene ; 31(22): 2783-93, 2012 May 31.
Article in English | MEDLINE | ID: mdl-21963850

ABSTRACT

Tyrosine kinase receptors have an essential role in various aspects of tumor progression. In particular, epidermal growth factor receptor (EGFR) and its ligands have been implicated in the growth and dissemination of a wide array of human carcinomas. Here, we describe an EGFR-mediated signaling pathway that regulates human pancreatic carcinoma cell invasion and metastasis, yet does not influence the growth of primary tumors. In fact, ligation/activation of EGFR induces Src-dependent phosphorylation of two critical tyrosine residues of p130CAS, leading to the assembly of a Crk-associated substrate (CAS)/Nck1 complex that promotes Ras-associated protein-1 (Rap1) signaling. Importantly, GTP loading of Rap1 is specifically required for pancreatic carcinoma cell migration on vitronectin but not on collagen. Furthermore, Rap1 activation is required for EGFR-mediated metastasis in vivo without impacting primary tumor growth. These findings identify a molecular pathway that promotes the invasive/metastatic properties of human pancreatic carcinomas driven by EGFR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , ErbB Receptors/metabolism , Oncogene Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/secondary , Telomere-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Chick Embryo , Humans , Immunoenzyme Techniques , Immunoprecipitation , Neoplasm Metastasis , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Shelterin Complex , Telomere-Binding Proteins/antagonists & inhibitors , Telomere-Binding Proteins/genetics
5.
Oncogene ; 28(40): 3551-62, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19668227

ABSTRACT

Microtubule-perturbing drugs have become front-line chemotherapeutics, inducing cell-cycle crisis as a major mechanism of action. However, these agents show pleiotropic effects on cells and can induce apoptosis through other means. Paclitaxel, a microtubule-stabilizing agent, induces a caspase-dependent apoptosis, although the precise mechanism(s) remain unclear. Here, we used genetic approaches to evaluate the role of caspase 8 in paclitaxel-mediated apoptosis. We observed that caspase 8-expressing cells are more sensitive to paclitaxel than caspase 8-deficient cells. Mechanistically, caspase 8 was found associated with microtubules, and this interaction increased after paclitaxel treatment. The prodomains death effector domains (DEDs) of caspase 8 were sufficient for interaction with microtubules, but the caspase 8 holoprotein was required for apoptosis. DED-only forms of caspase 8 were found in both primary and tumor cell lines, associating with perinuclear microtubules and the centrosome. Microtubule association, and paclitaxel sensitivity, depends on a critical lysine (K156) within a microtubule-binding motif (KLD) in DED-b of caspase 8. The results show an unexpected pathway of apoptosis mediated by caspase 8.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspase 8/physiology , Microtubules/physiology , Neuroblastoma/drug therapy , Paclitaxel/pharmacology , Caspase 8/chemistry , Cell Line, Tumor , Centrosome/physiology , Humans , Neuroblastoma/pathology , Protein Structure, Tertiary
6.
Oncogene ; 27(48): 6285-98, 2008 Oct 20.
Article in English | MEDLINE | ID: mdl-18931694

ABSTRACT

The extracellular matrix (ECM) acts both as a physical scaffold for cells and as a repository for growth factors. Moreover, ECM structure and physical-chemical properties convey precise information to cells that profoundly influences their biology by interactions with cell surface receptors termed integrins. During angiogenesis, the perivascular ECM plays a critical role in determining the proliferative, invasive and survival responses of the local vascular cells to the angiogenic growth factors. Dynamic changes in both the ECM and the local vascular cells act in concert to regulate new blood vessel growth. The digestion of ECM components by proteolysis is critical for the invasive capacity of endothelial cells, but also creates ECM fragments, which antagonize the mechanosensory function of integrins, and can be apoptogenic. Here, we discuss the roles of integrins in modulating cellular responses to a changing ECM, in particular the regulation of survival and invasion among invasive endothelial cells.


Subject(s)
Extracellular Matrix/physiology , Neovascularization, Physiologic/physiology , Animals , Apoptosis , Cell Adhesion , Cell Survival , Humans , Integrins/physiology , Signal Transduction
7.
Oncogene ; 27(10): 1439-48, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-17828307

ABSTRACT

Signals from fibronectin-binding integrins promote neural crest cell motility during development in part through protein-tyrosine kinase (PTK) activation. Neuroblastoma (NB) is a neural crest malignancy with high metastatic potential. We find that alpha4 and alpha5 integrins are present in late-stage NB tumors and cell lines derived thereof. To determine the signaling connections promoting either alpha4beta1- or alpha5beta1-initiated NB cell motility, pharmacological, dominant negative and short-hairpin RNA (shRNA) inhibitory approaches were undertaken. shRNA knockdown revealed that alpha5beta1-stimulated NB motility is dependent upon focal adhesion kinase (FAK) PTK, Src PTK and p130Cas adapter protein expression. Cell reconstitution showed that FAK catalytic activity is required for alpha5beta1-stimulated Src activation in part through direct FAK phosphorylation of Src at Tyr-418. Alternatively, alpha4beta1-stimulated NB cell motility is dependent upon Src and p130Cas but FAK is not essential. Catalytically inactive receptor protein-tyrosine phosphatase-alpha overexpression inhibited alpha4beta1-stimulated NB motility and Src activation consistent with alpha4-regulated Src activity occurring through Src Tyr-529 dephosphorylation. In alpha4 shRNA-expressing NB cells, alpha4beta1-stimulated Src activation and NB cell motility were rescued by wild type but not cytoplasmic domain-truncated alpha4 re-expression. These studies, supported by results using reconstituted fibroblasts, reveal that alpha4beta1-mediated Src activation is mechanistically distinct from FAK-mediated Src activation during alpha5beta1-mediated NB migration and support the evaluation of inhibitors to alpha4, Src and FAK in the control of NB tumor progression.


Subject(s)
Cell Movement/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alpha4beta1/physiology , Integrin alpha5beta1/physiology , Neuroblastoma/metabolism , Proto-Oncogene Proteins pp60(c-src)/physiology , Enzyme Activation/physiology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Integrin alpha4beta1/biosynthesis , Integrin alpha4beta1/genetics , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/genetics , Neuroblastoma/enzymology , Neuroblastoma/pathology , Protein Structure, Tertiary/physiology , Tumor Cells, Cultured
8.
Oncogene ; 25(44): 5969-84, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16682956

ABSTRACT

Elevated focal adhesion kinase (FAK) expression occurs in advanced cancers, yet a signaling role for FAK in tumor progression remains undefined. Here, we suppressed FAK activity in 4T1 breast carcinoma cells resulting in reduced FAK Y925 phosphorylation, Grb2 adaptor protein binding to FAK, and signaling to mitogen-activated protein (MAP) kinase (MAPK). Loss of a FAK-Grb2-MAPK linkage did not affect 4T1 cell proliferation or survival in culture, yet FAK inhibition reduced vascular endothelial growth factor (VEGF) expression and resulted in small avascular tumors in mice. This FAK-Grb2-MAPK linkage was essential in promoting angiogenesis as reconstitution experiments using Src-transformed FAK-null fibroblasts revealed that point mutations affecting FAK catalytic activity (R454) or Y925 phosphorylation (F925) disrupted the ability of FAK to promote MAPK- and VEGF-associated tumor growth. Notably, in both FAK-inhibited 4T1 and Src-transformed FAK-null cells, constitutively activated (CA) mitogen-activated protein kinase kinase 1 (MEK1) restored VEGF production and CA-MEK1 or added VEGF rescued tumor growth and angiogenesis. These studies provide the first biological support for Y925 FAK phosphorylation and define a novel role for FAK activity in promoting a MAPK-associated angiogenic switch during tumor progression.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mammary Neoplasms, Animal/enzymology , Neovascularization, Pathologic/enzymology , Tyrosine/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Carcinoma/blood supply , Carcinoma/enzymology , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Clone Cells , Female , Focal Adhesion Protein-Tyrosine Kinases/deficiency , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Mammary Neoplasms, Animal/blood supply , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein-Tyrosine Kinases/physiology , Signal Transduction/genetics
9.
Cell Death Differ ; 12(8): 1021-30, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15933741

ABSTRACT

In the absence of their cognate ligand, dependence receptors trigger programmed cell death. This function is the defining feature of dependence receptors, which include members of several different protein families. The integrins are a family of heterodimeric receptors for extracellular matrix (ECM) proteins, mediating cell anchorage and migration. Integrins share characteristics with dependence receptors, and integrin binding to substrate ECM ligands is essential for cell survival. Although integrins do not conform in all characteristics to the established definitions of dependence receptors, alterations in the expression of integrins and their ligands during physiological and pathological events, such as wound healing, angiogenesis and tumorigenesis, do regulate cell fate in a ligand-dependent manner. This biosensory function of integrins fits well with our current concept of dependence receptor action, and thus integrins may rightly be considered to comprise a distinct subclass of dependence receptor.


Subject(s)
Apoptosis/physiology , Integrins/physiology , Animals , Extracellular Matrix/physiology , Humans , Ligands
10.
Curr Top Dev Biol ; 64: 207-38, 2004.
Article in English | MEDLINE | ID: mdl-15563949

ABSTRACT

The growth of new blood vessels is a dynamic yet highly regulated process that depends on coordinated signaling by growth factor and cell adhesion receptors. As part of the molecular program regulating angiogenesis, endothelial cells acquire a proliferative and invasive phenotype but also show increased susceptibility to apoptotic stimuli. Integrins are the principle adhesion receptors used by endothelial cells to interact with their extracellular microenvironment, and integrin-mediated interactions play a critical role in regulating cell proliferation, migration, and survival. Alterations in the repertoire and?or activity of integrins, as well as the availability and structural property of their ligands, regulate the vascular cell during the growth or repair of blood vessels.


Subject(s)
Endothelial Cells/metabolism , Integrins , Neovascularization, Physiologic , Animals , Blood Vessels/anatomy & histology , Blood Vessels/physiology , Cell Survival , Clinical Trials as Topic , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Fibrin/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism
11.
J Cell Biol ; 155(3): 459-70, 2001 Oct 29.
Article in English | MEDLINE | ID: mdl-11684710

ABSTRACT

Integrin-mediated adhesion promotes cell survival in vitro, whereas integrin antagonists induce apoptosis of adherent cells in vivo. Here, we demonstrate that cells adherent within a three-dimensional extracellular matrix undergo apoptosis due to expression of unligated integrins, the beta subunit cytoplasmic domain, or its membrane proximal sequence KLLITIHDRKEF. Integrin-mediated death requires initiator, but not stress, caspase activity and is distinct from anoikis, which is caused by the loss of adhesion per se. Surprisingly, unligated integrin or beta integrin tails recruit caspase-8 to the membrane, where it becomes activated in a death receptor-independent manner. Integrin ligation disrupts this integrin-caspase containing complex and increases survival, revealing an unexpected role for integrins in the regulation of apoptosis and tissue remodeling.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Caspases/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Antigens, CD/genetics , Binding Sites , COS Cells , Caspase 8 , Caspase 9 , Caspase Inhibitors , Cell Adhesion , Cell Membrane/metabolism , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/metabolism , Integrin alpha5 , Integrin beta1/genetics , Integrin beta1/metabolism , Integrin beta3 , Platelet Membrane Glycoproteins/genetics , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Receptors, Vitronectin/metabolism
12.
J Virol ; 75(11): 5405-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11333925

ABSTRACT

The human embryonic kidney (HEK293) cell line, commonly used for recombinant adenovirus (Ad) propagation, does not express the Ad coreceptor alpha(v)beta3 or alpha(v)beta5 integrins, yet these cells are efficiently infected by Ad vectors. Here we demonstrate that Ad binds to HEK293 cells via the fiber receptor CAR and is subsequently internalized via interaction with integrin alpha(v)beta1. Function-blocking antibodies directed against alpha(v) or beta1, but not beta3, beta5, or alpha5, integrin subunits block Ad infection and viral endocytosis. Therefore, alpha(v)beta1 serves as a coreceptor for Ad infection, and the lack of beta3 and/or beta5 but the relatively high expression of alpha(v)beta1 integrins on certain tumor cell types may explain why these cells are readily transduced by Ad vectors.


Subject(s)
Adenoviridae/physiology , Capsid Proteins , Integrins/metabolism , Receptors, Virus/metabolism , Receptors, Vitronectin , Capsid/metabolism , Cell Adhesion , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Humans
13.
Can J Physiol Pharmacol ; 79(1): 18-24, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11201497

ABSTRACT

The effects of early-stage diabetes mellitus and uninephrectomy on the renal tubule transport of amantadine were investigated. Kidney tubules were isolated from streptozotocin-induced diabetic (+/- insulin treatment) uninephrectomized, and control male Sprague-Dawley rats. There were no differences in the Km of amantadine uptake in renal proximal and distal tubules for the imposed treatments compared with control values. Vmax for amantadine uptake in the proximal tubules of diabetic and uninephrectomized rats was higher than the respective control (P < 0.05). Vmax for insulin-treated diabetic rats was similar to control values but was lower than that for untreated diabetic rats (P < 0.05). Vmax for distal tubule uptake was not altered by any treatment. Structure-activity studies demonstrated that bicarbonate-dependent amantadine uptake was inhibited by glycolate and lactate, but not by propionate or alpha-, beta-, or gamma-hydroxybutyrate. Early stage streptozotocin-induced diabetes mellitus and uninephrectomy induced changes in the kidney that resulted in a similar selective increase in proximal tubule amantadine uptake. These data represent the first description that experimentally induced diabetes mellitus and uninephrectomy modulate the function of the renal tubule organic cation (amantadine) transport system. Both interventions represent potential models in which phenotypic modulation of the renal elimination of organic cationic drugs may be achieved and studied.


Subject(s)
Amantadine/pharmacokinetics , Diabetes Mellitus, Experimental/metabolism , Dopamine Agents/pharmacokinetics , Kidney Tubules/metabolism , Nephrectomy , Animals , Bicarbonates/metabolism , Carrier Proteins/metabolism , Energy Metabolism/physiology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Lactates/metabolism , Male , Rats , Rats, Sprague-Dawley
14.
Immunol Res ; 21(2-3): 83-8, 2000.
Article in English | MEDLINE | ID: mdl-10852105

ABSTRACT

The ability of immune cells to migrate and invade the extracellular matrix (ECM) is a central process involved in immunologic surveillance as well as inflammation. We have shown that interaction of cells with adhesive proteins or growth factors (chemokines) present in the ECM control cell migration/invasion through activation of mitogen-activated protein kinases ERK1 and ERK2 and molecular coupling of the adapter proteins p130CAS and c-CrkII. During cell migration, ERK and CAS/Crk coupling operate as distinct signaling pathways that facilitate actin-myosin motor assembly and actin membrane ruffles, respectively. Furthermore, activation of these signaling pathways protects cells from apoptosis during invasion of the ECM, which is necessary as migratory cells colonize foreign sites in the body.


Subject(s)
Cell Movement/immunology , Immunity, Cellular , Signal Transduction/immunology , Animals , Cell Adhesion/immunology , Extracellular Matrix/immunology , Humans , MAP Kinase Signaling System/immunology
15.
J Biol Chem ; 275(19): 14729-35, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10799562

ABSTRACT

The Crk-associated substrate, p130(CAS), has been implicated in the regulation of the actin cytoskeleton following ligation of cell integrins with the extracellular matrix. Integrin-mediated cell adhesion involves p130(CAS) association with focal adhesion kinase (p125(FAK)). Internalization/cell entry of type 2 and type 5 adenoviruses (Ad) is also mediated by alpha(v) integrins. However, expression of dominant negative forms of p125(FAK) does not alter virus entry, and Ad entry occurs normally in p125(FAK)-deficient fibroblasts. We now provide evidence that Ad internalization, a process which is mediated by alpha(v) integrins, also requires p130(CAS) and phosphatidylinositol-3-OH kinase (PI 3-kinase). Ad induces p130(CAS) phosphorylation and inhibition of p130(CAS) phosphorylation by tyrphostin and genistein, or expression of the substrate domain deleted p130(CAS) blocks Ad internalization. p130(CAS) was also found to associate with the p85 subunit of PI 3-kinase through its proline-rich domain during virus internalization and expression of p130(CAS) containing a deleted proline-rich domain (PRD) inhibited adenovirus cell entry. We showed further that the RPLPSPP motif in the proline-rich region of p130(CAS) interacts with the SH3 domain of p85/PI 3-kinase. These studies reveal the molecular basis by which p130(CAS) coordinates the signaling pathways involved in integrin-mediated Ad endocytosis.


Subject(s)
Adenoviridae/physiology , Membrane Fusion , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Proteins , Receptors, Vitronectin , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Crk-Associated Substrate Protein , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Integrins/physiology , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Retinoblastoma-Like Protein p130
16.
Oncogene ; 19(15): 1915-23, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773881

ABSTRACT

alpha(v) integrins have been shown to play an important role in epithelial-derived cell migration, cell growth and tumor invasion/metastasis, however their role on cells of hematopoietic origin is less clear. Epstein-Barr virus (EBV), a human herpesvirus associated with several lymphoproliferative disorders in man, induces expression of alpha(v) integrins on transformed B lymphocytes. In the studies reported here, we show that EBV infection increases alpha(v), beta3 and beta5 integrin subunit mRNAs as well as upregulates the expression of the alphavbeta3 integrin protein on human B cells. Among the nine different EBV proteins expressed in latently infected B cells (nuclear and plasma membrane-associated), only LMP1, LMP2A and EBNA2 were shown to selectively transactivate the alpha(v) integrin promoter. Treatment of EBV-transformed B cells with alpha(v) antisense oligonucleotides specifically reduced cell surface expression of alpha(v) integrins, inhibited cell growth in low serum, reduced cell invasion in matrigels and decreased expression of metalloprotease, MMP9. These studies indicate that alpha(v) integrins play a significant role in EBV-induced B-lymphocyte proliferation and invasion. Strategies to interfere with alphav integrin expression and/or function may therefore be of potential value in the treatment of EBV-associated lymphoproliferative disorders.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/physiology , Epstein-Barr Virus Nuclear Antigens , Herpesvirus 4, Human/genetics , B-Lymphocytes/metabolism , Cell Division , Cell Line, Transformed , Gene Expression Regulation, Viral , Humans , Integrin alphaV , Integrins/metabolism , Neoplasm Invasiveness , Oligonucleotides, Antisense/pharmacology , Transcriptional Activation , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism
17.
Can J Physiol Pharmacol ; 77(6): 407-13, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10537226

ABSTRACT

Eight representative beta-adrenoreceptor blocking drugs, acebutolol, atenolol, labetalol, metoprolol, nadolol, pindolol, propranolol, and timolol, were studied in vitro with respect to their potential to block energy-dependent uptake of [3H]amantadine into proximal and distal rat renal tubule fragments in the presence and absence of bicarbonate. Five of the eight beta-adrenoreceptor blockers showed a dose-dependent inhibition of renal tubule accumulation of amantadine: labetalol, metoprolol, pindolol, propranolol, and timolol. Labetalol was the only beta-adrenoreceptor blocker with greater inhibitory potency in phosphate-based buffer than in bicarbonate-based buffer. Propranolol was the most potent inhibitor of renal tubule amantadine accumulation with IC50 values of 15 +/- 10 and 31 +/- 11 microM for proximal and distal tubule fragments, respectively, in a bicarbonate-based buffer environment. Inhibition in a phosphate-based buffer was less potent only in proximal tubules, with an IC50 of 76 +/- 30 microM. Kinetic studies of propranolol inhibition of amantadine uptake were consistent with both uncompetitive and competitive inhibition mechanisms in bicarbonate-based buffer in both proximal and distal renal tubule segments. There was no chiral preference between the R and S forms of propranolol for the inhibitory effects observed. These data suggest that there is potential for selection among the beta-adrenoreceptor blocking drugs to minimize or restrict the inhibition of amantadine energy-dependent uptake at the organic cation ion uptake sites characterized by amantadine in the presence and absence of bicarbonate.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Amantadine/pharmacokinetics , Kidney Tubules/drug effects , Animals , Bicarbonates/pharmacology , Dose-Response Relationship, Drug , Kidney Tubules/metabolism , Male , Rats , Rats, Sprague-Dawley , Stereoisomerism
18.
Braz J Med Biol Res ; 32(5): 573-81, 1999 May.
Article in English | MEDLINE | ID: mdl-10412569

ABSTRACT

Rheumatoid arthritis (RA) is a chronic debilitating disease characterized by distinct autoimmune, inflammatory and fibrovascular components which lead to synovial proliferation and joint destruction. However, existing treatments specifically target only autoimmune and inflammatory components despite the fact that neovascularization of the inflamed synovium is a hallmark of rheumatoid arthritis. Angiogenesis may contribute to synovial growth, leukocyte recruitment and tissue remodeling, thus potentiating disease progression. Although no therapies currently target angiogenesis, several existing therapies have anti-angiogenic activity. Recent advances in anti-angiogenic strategies in oncology, including the identification of integrin alpha v beta 3 as a crucial effector of angiogenesis, suggest a means to assess the role of angiogenesis in rheumatoid arthritis. Synovial endothelial cells have been shown to express integrin alpha v beta 3, suggesting that these cells may be targeted for angiogenesis inhibition. Prior studies in rat arthritis models have shown benefit after the addition of broad spectrum integrin antagonists. However, formal assessment of integrin-targeted anti-angiogenic activity is now underway. These controlled studies will be important in assessing the efficacy of therapies which target angiogenesis in RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Neovascularization, Pathologic , Animals , Arthritis, Rheumatoid/therapy , Endothelial Growth Factors , Endothelium, Vascular/pathology , Receptors, Vitronectin/immunology , Synovial Membrane/blood supply
19.
J Biol Chem ; 274(31): 21609-16, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419468

ABSTRACT

Integrin alpha(V)beta(3) mediates diverse responses in vascular cells, ranging from cell adhesion, migration, and proliferation to uptake of adenoviruses. However, the extent to which alpha(V)beta(3) is regulated by changes in receptor conformation (affinity), receptor diffusion/clustering (avidity), or post-receptor events is unknown. Affinity regulation of the related integrin, alpha(IIb)beta(3), has been established using a monovalent ligand-mimetic antibody, PAC1 Fab. To determine the role of affinity modulation of alpha(V)beta(3), a novel monovalent ligand-mimetic antibody (WOW-1) was created by replacing the heavy chain hypervariable region 3 of PAC1 Fab with a single alpha(V) integrin-binding domain from multivalent adenovirus penton base. Both WOW-1 Fab and penton base bound selectively to activated alpha(V)beta(3), but not to alpha(IIb)beta(3), in receptor and cell binding assays. alpha(V)beta(3) affinity varied with the cell type. Unstimulated B-lymphoblastoid cells bound WOW-1 Fab poorly (apparent K(d) = 2.4 microM), but acute stimulation with phorbol 12-myristate 13-acetate increased receptor affinity >30-fold (K(d) = 80 nM), with no change in receptor number. In contrast, alpha(V)beta(3) in melanoma cells was constitutively active, but ligand binding could be suppressed by overexpression of beta(3) cytoplasmic tails. Up-regulation of alpha(V)beta(3) affinity had functional consequences in that it increased cell adhesion and spreading and promoted adenovirus-mediated gene transfer. These studies establish that alpha(V)beta(3) is subject to rapid regulated changes in affinity that influence the biological functions of this integrin.


Subject(s)
Receptors, Vitronectin/physiology , Adenoviridae , Animals , Antibody Affinity , B-Lymphocytes/immunology , CHO Cells , Cell Line , Cloning, Molecular , Cricetinae , Drosophila melanogaster , Fibrinogen/metabolism , Genetic Vectors , Humans , Immunoglobulin Fab Fragments/pharmacology , Kinetics , Ligands , Polymerase Chain Reaction , Receptors, Vitronectin/drug effects , Receptors, Vitronectin/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Up-Regulation
20.
J Cell Biol ; 144(4): 777-88, 1999 Feb 22.
Article in English | MEDLINE | ID: mdl-10037798

ABSTRACT

Lymphocytes accumulate within the extracellular matrix (ECM) of tumor, wound, or inflammatory tissues. These tissues are largely comprised of polymerized adhesion proteins such as fibrin and fibronectin or their fragments. Nonactivated lymphoid cells attach preferentially to polymerized ECM proteins yet are unable to attach to monomeric forms or fragments of these proteins without previous activation. This adhesion event depends on the appropriate spacing of integrin adhesion sites. Adhesion of nonactivated lymphoid cells to polymeric ECM components results in activation of the antigen receptor-associated Syk kinase that accumulates in adhesion-promoting podosomes. In fact, activation of Syk by antigen or agonists, as well as expression of an activated Syk mutant in lymphoid cells, facilitates their adhesion to monomeric ECM proteins or their fragments. These results reveal a cooperative interaction between signals emanating from integrins and antigen receptors that can serve to regulate stable lymphoid cell adhesion and retention within a remodeling ECM.


Subject(s)
Enzyme Precursors/metabolism , Extracellular Matrix/metabolism , Integrins/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Protein-Tyrosine Kinases/metabolism , Cell Adhesion/physiology , Cell Line , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Integrins/chemistry , Intracellular Signaling Peptides and Proteins , Ligands , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Syk Kinase , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...