Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 3: e437, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23190609

ABSTRACT

Necrostatin-1 (Nec-1) is widely used in disease models to examine the contribution of receptor-interacting protein kinase (RIPK) 1 in cell death and inflammation. We studied three Nec-1 analogs: Nec-1, the active inhibitor of RIPK1, Nec-1 inactive (Nec-1i), its inactive variant, and Nec-1 stable (Nec-1s), its more stable variant. We report that Nec-1 is identical to methyl-thiohydantoin-tryptophan, an inhibitor of the potent immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO). Both Nec-1 and Nec-1i inhibited human IDO, but Nec-1s did not, as predicted by molecular modeling. Therefore, Nec-1s is a more specific RIPK1 inhibitor lacking the IDO-targeting effect. Next, although Nec-1i was ∼100 × less effective than Nec-1 in inhibiting human RIPK1 kinase activity in vitro, it was only 10 times less potent than Nec-1 and Nec-1s in a mouse necroptosis assay and became even equipotent at high concentrations. Along the same line, in vivo, high doses of Nec-1, Nec-1i and Nec-1s prevented tumor necrosis factor (TNF)-induced mortality equally well, excluding the use of Nec-1i as an inactive control. Paradoxically, low doses of Nec-1 or Nec-1i, but not Nec -1s, even sensitized mice to TNF-induced mortality. Importantly, Nec-1s did not exhibit this low dose toxicity, stressing again the preferred use of Nec-1s in vivo. Our findings have important implications for the interpretation of Nec-1-based data in experimental disease models.


Subject(s)
Imidazoles/administration & dosage , Imidazoles/chemistry , Indoles/administration & dosage , Indoles/chemistry , Systemic Inflammatory Response Syndrome/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Therapy , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Species Specificity
2.
Curr Med Chem ; 18(15): 2257-62, 2011.
Article in English | MEDLINE | ID: mdl-21517753

ABSTRACT

Chronic inflammation underlies the basis for development and progression of cancers and a variety of other disorders, but what specifically defines its pathogenic nature remains largely undefined. Recent genetic and pharmacological studies in the mouse suggest that the immune modulatory enzyme indoleamine 2,3-dioxygenase (IDO), identified as an important mediator of immune escape in cancer, can also contribute to the development of pathology in the context of chronic inflammatory models of arthritis and allergic airway disease. IDO-deficient mice do not display spontaneous disorders of classical inflammation and small molecule inhibitors of IDO do not elicit generalized inflammatory reactions. Rather, in the context of a classical model of skin cancer that is promoted by chronic inflammation, or in models of inflammation-associated arthritis and allergic airway disease, IDO impairment can alleviate disease severity. Here we offer a survey of preclinical literature suggesting that IDO functions as a modifier of inflammatory states rather than simply as a suppressor of immune function. We propose that IDO induction in a chronically inflamed tissue may shape the inflammatory state to support, or in some cases retard, pathogenesis and disease severity.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Inflammation/enzymology , Inflammation/immunology , Neoplasms/enzymology , Neoplasms/immunology , Animals , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/immunology , Asthma/enzymology , Asthma/immunology , Humans , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/immunology
3.
Oncogene ; 27(58): 7223-34, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-18806825

ABSTRACT

The pathophysiological mechanisms that drive the development and progression of epithelial ovarian cancer remain obscure. Recently, we identified TCEAL7 as a transcriptional regulatory protein often downregulated in epithelial ovarian cancer. However, the biological significance of such downregulation in cancer is not currently known. Here, we show that TCEAL7 is downregulated frequently in many human cancers and that in immortalized human ovarian epithelial cells this event promotes anchorage-independent cell growth. Mechanistic investigations revealed that TCEAL7 associates with cyclin D1 promoter containing Myc E-box sequence and transcriptionally represses cyclin D1 expression. Moreover, downregulation of TCEAL7 promotes DNA-binding activity of Myc-Max, and upregulates the promoter activity of c-Myc-target gene, ornithine decarboxylase (ODC), whereas enhanced expression of TCEAL7 inhibits Myc-induced promoter activity of ODC. Our findings suggest that TCEAL7 may restrict ovarian epithelial cell transformation by limiting Myc activity. These results also suggest a potential, alternative mechanism by which c-Myc activity may be deregulated in cancer by the downregulation of TCEAL7.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Biological Phenomena , Cell Line, Transformed , Cell Line, Tumor , Female , Genes, myc , HeLa Cells , Humans , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics
4.
Oncogene ; 27(28): 3889-900, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18317452

ABSTRACT

Immune escape is a critical gateway to malignancy. The emergence of this fundamental trait of cancer represents the defeat of immune surveillance, a potent, multi-armed and essential mode of cancer suppression that may influence the ultimate clinical impact of an early stage tumor. Indeed, immune escape may be a central modifier of clinical outcomes, by affecting tumor dormancy versus progression, licensing invasion and metastasis and impacting therapeutic response. Although relatively little studied until recently, immune suppression and escape in tumors are now hot areas with clinical translation of several new therapeutic agents already under way. The interconnections between signaling pathways that control immune escape and those that control proliferation, senescence, apoptosis, metabolic alterations, angiogenesis, invasion and metastasis remain virtually unexplored, offering rich new areas for investigation. Here, an overview of this area is provided with a focus on the tryptophan catabolic enzyme indoleamine 2,3-dioxygenase (IDO) and its recently discovered relative IDO2 that are implicated in suppressing T-cell immunity in normal and pathological settings including cancer. Emerging evidence suggests that during cancer progression activation of the IDO pathway might act as a preferred nodal modifier pathway for immune escape, for example analogous to the PI3K pathway for survival or the VEGF pathway for angiogenesis. Small molecule inhibitors of IDO and IDO2 heighten chemotherapeutic efficacy in mouse models of cancer in a nontoxic fashion and an initial lead compound entered phase I clinical trials in late 2007. New modalities in this area offer promising ways to broaden the combinatorial attack on advanced cancers, where immune escape mechanisms likely provide pivotal support.


Subject(s)
Gene Expression Regulation, Neoplastic , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Neoplasms/immunology , Neoplasms/pathology , Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Disease Progression , Humans , Immunotherapy/methods , Models, Biological , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polymorphism, Genetic , Signal Transduction , Tumor Suppressor Proteins/metabolism
5.
Oncogene ; 27(20): 2851-7, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18026137

ABSTRACT

Agents that interfere with tumoral immune tolerance may be useful to prevent or treat cancer. Brassinin is a phytoalexin, a class of natural products derived from plants that includes the widely known compound resveratrol. Brassinin has been demonstrated to have chemopreventive activity in preclinical models but the mechanisms underlying its anticancer properties are unknown. Here, we show that brassinin and a synthetic derivative 5-bromo-brassinin (5-Br-brassinin) are bioavailable inhibitors of indoleamine 2,3-dioxygenase (IDO), a pro-toleragenic enzyme that drives immune escape in cancer. Like other known IDO inhibitors, both of these compounds combined with chemotherapy to elicit regression of autochthonous mammary gland tumors in MMTV-Neu mice. Furthermore, growth of highly aggressive melanoma isograft tumors was suppressed by single agent treatment with 5-Br-brassinin. This response to treatment was lost in athymic mice, indicating a requirement for active host T-cell immunity, and in IDO-null knockout mice, providing direct genetic evidence that IDO inhibition is essential to the antitumor mechanism of action of 5-Br-brassinin. The natural product brassinin thus provides the structural basis for a new class of compounds with in vivo anticancer activity that is mediated through the inhibition of IDO.


Subject(s)
Antineoplastic Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/drug therapy , Thiocarbamates/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Mammary Neoplasms, Experimental/enzymology , Melanoma, Experimental/enzymology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Structure-Activity Relationship
6.
Oncogene ; 25(9): 1281-9, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16247449

ABSTRACT

The small GTPase RhoB suppresses cancer in part by limiting cell proliferation. However, the mechanisms it uses to achieve this are poorly understood. Recent studies link RhoB to trafficking of Akt, which through its regulation of glycogen synthase kinase-3 (GSK-3) has an important role in controlling the stability of the c-Myc oncoprotein. c-Myc stabilization may be a root feature of human tumorigenesis as it phenocopies an essential contribution of SV40 small T antigen in human cell transformation. In this study we show that RhoB directs efficient turnover of c-Myc in established or transformed mouse fibroblasts and that the attenuation of RhoB which occurs commonly in human cancer is a sufficient cause to elevate c-Myc levels. Increased levels of c-Myc elicited by RhoB deletion increased the proliferation of nullizygous cells, whereas restoring RhoB in null cells decreased the stability of c-Myc and restrained cell proliferation. Mechanistic analyses indicated that RhoB facilitated nuclear accumulation of GSK-3 and GSK-3-mediated phosphorylation of c-Myc T58, the critical site for ubiquitination and degradation of c-Myc. RhoB deletion restricted nuclear localization of GSK-3, reduced T58 phosphorylation, and stabilized c-Myc. These effects were not associated with changes in phosphorylation or localization of Akt, however, differences were observed in phosphorylation and localization of the GSK-3 regulatory Akt-related kinase, serum- and glucocorticoid-inducible protein kinase (SGK). The ability of RhoB to support GSK-3-dependent turnover of c-Myc offers a mechanism by which RhoB acts to limit the proliferation of neoplastically transformed cells.


Subject(s)
Glycogen Synthase Kinase 3/pharmacokinetics , Neoplasms/physiopathology , Proto-Oncogene Proteins c-myc/metabolism , rhoB GTP-Binding Protein/physiology , Animals , Cell Culture Techniques , Cell Nucleus/enzymology , Cell Proliferation , Cell Transformation, Neoplastic , Fibroblasts , Humans , Mice , Neoplasms/genetics , Phenotype , Transformation, Genetic
7.
Histol Histopathol ; 21(2): 213-8, 2006 02.
Article in English | MEDLINE | ID: mdl-16329046

ABSTRACT

RhoB is a mainly endosomal small GTPase that regulates actin organization and vesicle trafficking. Expression of RhoB is elevated rapidly by many stimuli, including growth factors, cytokines, and genotoxic stress. In cancer, RhoB can limit cell proliferation, survival, invasion, and metastasis, and during malignant progression its levels are attenuated commonly. In support of its role as a negative modifier of cancer progression, targeted deletion of RhoB in mice can increase tumor formation initiated by Ras mutation. How RhoB acts to suppress different aspects of cancer pathophysiology has emerged as a question of significant interest.


Subject(s)
Neoplasms/physiopathology , rhoB GTP-Binding Protein/physiology , Actins/physiology , Animals , Cell Death , Cell Proliferation , Endocytosis , Farnesyltranstransferase/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Mutation , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction , rhoB GTP-Binding Protein/genetics
9.
Mol Cell Biol ; 21(20): 6906-12, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564874

ABSTRACT

RhoB is an endosomal small GTPase that is implicated in the response to growth factors, genotoxic stress, and farnesyltransferase inhibitors. To gain insight into its physiological functions we examined the consequences of homozygous gene deletion in the mouse. Loss of RhoB did not adversely affect mouse development, fertility, or wound healing. However, embryo fibroblasts cultured in vitro exhibited a defect in motility, suggesting that RhoB has a role in this process that is conditional on cell stress. Neoplastic transformation by adenovirus E1A and mutant Ras yielded differences in cell attachment and spreading that were not apparent in primary cells. In addition, transformed -/- cells displayed altered actin and proliferative responses to transforming growth factor beta. A negative modifier role in transformation was suggested by the increased susceptibility of -/- mice to 7,12-dimethylbenz[a]anthracene-induced skin carcinogenesis and by the increased efficiency of intraperitoneal tumor formation by -/- cells. Our findings suggest that RhoB is a negative regulator of integrin and growth factor signals that are involved in neoplastic transformation and possibly other stress or disease states.


Subject(s)
Genetic Predisposition to Disease , Growth Substances/metabolism , Neoplasms/metabolism , Signal Transduction , rhoB GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/physiology , 9,10-Dimethyl-1,2-benzanthracene , Actins/metabolism , Animals , Blotting, Western , Carcinogens , Cell Adhesion , Cell Line, Transformed , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibronectins/metabolism , Gene Deletion , Homozygote , Mice , Mice, Transgenic , Models, Genetic , Neoplasms/genetics , Skin Neoplasms/chemically induced , Time Factors , Transforming Growth Factor beta/pharmacology
10.
Proc Natl Acad Sci U S A ; 98(11): 6192-7, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11353846

ABSTRACT

The effect of neoplastic transformation on the response to genotoxic stress is of significant clinical interest. In this study, we offer genetic evidence that the apoptotic response of neoplastically transformed cells to DNA damage requires RhoB, a member of the Rho family of actin cytoskeletal regulators. Targeted deletion of the rhoB gene did not affect cell cycle arrest in either normal or transformed cells after exposure to doxorubicin or gamma irradiation, but rendered transformed cells resistant to apoptosis. This effect was specific insofar as rhoB deletion did not affect apoptotic susceptibility to agents that do not damage DNA. However, rhoB deletion also affected apoptotic susceptibility to Taxol, an agent that disrupts microtubule dynamics. We have demonstrated that RhoB alteration mediates the proapoptotic and antineoplastic effects of farnesyltransferase inhibitors, and we show here that RhoB alteration is also crucial for farnesyltransferase inhibitors to sensitize neoplastic cells to DNA damage-induced cell death. We found RhoB to be an important determinant of long-term survival in vitro and tumor response in vivo after gamma irradiation. Our findings identify a pivotal role for RhoB in the apoptotic response of neoplastic cells to DNA damage at a novel regulatory point that may involve the actin cytoskeleton.


Subject(s)
Apoptosis , DNA Damage , rhoB GTP-Binding Protein/physiology , Alkyl and Aryl Transferases/antagonists & inhibitors , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Farnesyltranstransferase , Fibroblasts/cytology , Gene Targeting , Mice , Paclitaxel/pharmacology , rhoB GTP-Binding Protein/genetics
11.
Cancer Res ; 61(7): 3151-6, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306501

ABSTRACT

The Bin1 gene encodes a c-Myc-interacting adapter protein with tumor suppressor and cell death properties. In this study, we offer evidence that Bin1 participates in a mechanism through which c-Myc activates programmed cell death in transformed primary chick or rat cells. Antisense or dominant inhibitory Bin1 genes did not affect the ability of c-Myc to drive proliferation or transformation, but they did reduce the susceptibility of cells to c-Myc-induced apoptosis. Protein-protein interaction was implicated, suggesting that Bin1 mediates a death or death sensitization signal from c-Myc. Our findings offer direct support for the "dual signal" model of Myc apoptotic function, based on interactions with a binding protein. Loss of Bin1 in human tumors may promote malignant progression in part by helping to stanch the death penalty associated with c-Myc activation.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Cell Transformation, Neoplastic , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chick Embryo , Epithelial Cells/pathology , Epithelial Cells/physiology , Fibroblasts/pathology , Gene Expression Regulation , Genes, myc/genetics , Genes, myc/physiology , Humans , Kidney/pathology , Kidney/physiology , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
12.
J Biol Chem ; 276(24): 21670-7, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11274158

ABSTRACT

The BAR adaptor proteins encoded by the RVS167 and RVS161 genes from Saccharomyces cerevisiae form a complex that regulates actin, endocytosis, and viability following starvation or osmotic stress. In this study, we identified a human homolog of RVS161, termed BIN3 (bridging integrator-3), and a Schizosaccharomyces pombe homolog of RVS161, termed hob3+ (homolog of Bin3). In human tissues, the BIN3 gene was expressed ubiquitously except for brain. S. pombe cells lacking Hob3p were often multinucleate and characterized by increased amounts of calcofluor-stained material and mislocalized F-actin. For example, while wild-type cells localized F-actin to cell ends during interphase, hob3Delta mutants had F-actin patches distributed randomly around the cell. In addition, medial F-actin rings were rarely found in hob3Delta mutants. Notably, in contrast to S. cerevisiae rvs161Delta mutants, hob3Delta mutants showed no measurable defects in endocytosis or response to osmotic stress, yet hob3+ complemented the osmosensitivity of a rvs161Delta mutant. BIN3 failed to rescue the osmosensitivity of rvs161Delta, but the actin localization defects of hob3Delta mutants were completely rescued by BIN3 and partially rescued by RVS161. These findings suggest that hob3+ and BIN3 regulate F-actin localization, like RVS161, but that other roles for this gene have diverged somewhat during evolution.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins , Fungal Proteins/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Saccharomyces cerevisiae Proteins , Schizosaccharomyces/genetics , Transcription, Genetic , Actins/analysis , Amino Acid Sequence , Brain/metabolism , Cell Line , DNA, Complementary , Fungal Proteins/chemistry , Gene Library , Genetic Complementation Test , Humans , Kinetics , Microfilament Proteins/chemistry , Molecular Sequence Data , Organ Specificity , RNA, Messenger/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces/metabolism , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid
13.
Histol Histopathol ; 16(1): 269-75, 2001 01.
Article in English | MEDLINE | ID: mdl-11193202

ABSTRACT

A long-standing goal in cancer research is to identify cellular functions that have selective roles in regulating neoplastic pathophysiology. Farnesyl-transferase inhibitors (FTIs) are a novel class of cancer chemotherapeutics which have little effect on normal cell physiology but which inhibit or reverse malignant cell phenotypes. FTIs were originally developed as a strategy to inhibit oncogenic Ras, the activity of which depends upon posttranslational farnesylation. However, recent work indicates the antineoplastic effects of FTIs are not linked to Ras inhibition but instead to alteration of RhoB, a small GTPase of the Rho family of cytoskeletal regulators that controls trafficking of cell surface receptors. Rho proteins integrate signals from integrins and cytokine receptors with cell shape via the actin cytoskeleton. A connection between FTIs and Rho alteration is interesting given that histological differences have long been used to define clinical cancer. RhoB is dispensable for normal cell growth and differentiation in mice. Thus, research into the antineoplastic effects of FTIs has led to the identification of a function(s) that is unnecessary for normal cell physiology but crucial for controlling malignant phenotypes.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/pathology , rhoB GTP-Binding Protein/physiology , Animals , Farnesyltranstransferase , Humans , Neoplasms/enzymology , rhoB GTP-Binding Protein/drug effects
14.
Nat Rev Cancer ; 1(2): 162-8, 2001 11.
Article in English | MEDLINE | ID: mdl-11905808

ABSTRACT

RhoB is a small GTPase that regulates actin organization and vesicle transport. It is required for signalling apoptosis in transformed cells that are exposed to farnesyltransferase inhibitors, DNA-damaging agents or taxol. Genetic analysis in mice indicates that RhoB is dispensable for normal cell physiology, but that it has a suppressor or negative modifier function in stress-associated processes, including cancer.


Subject(s)
Actins/metabolism , Apoptosis/physiology , Neoplasms/metabolism , rhoB GTP-Binding Protein/physiology , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion/physiology , Cell Division/physiology , DNA Damage , Enzyme Induction , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Guanosine Triphosphate/metabolism , Humans , Mice , Multigene Family , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Paclitaxel/pharmacology , Protein Prenylation/drug effects , Protein Processing, Post-Translational/drug effects , Signal Transduction/physiology , Stress, Physiological/genetics , Stress, Physiological/metabolism , ras Proteins/antagonists & inhibitors
15.
Expert Opin Investig Drugs ; 10(12): 2105-16, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11772308

ABSTRACT

Farnesyltransferase (FT) inhibitors (FTIs) are among the first wave of signal transduction inhibitors to be clinically tested for antitumour properties. FTIs were designed to attack Ras oncoproteins, the function of which depends upon post-translational modification by farnesyl isoprenoid. Extensive preclinical studies have demonstrated that FTIs compromise neoplastic transformation and tumour growth. In preclinical models, FTIs display limited effects on normal cell physiology and in Phase I human trials FTIs have been largely well tolerated. Exactly how FTIs selectively target cancer cells has emerged as an important question, one which has become more pressing with the somewhat disappointing results from initial Phase II efficacy trials. Although FTI development was predicated on Ras inhibition, it has become clear that the drugs' antineoplastic properties are based to a large degree on altering the prenylation and function of proteins other than Ras. One key candidate that has emerged is RhoB, an endosomal protein that has been implicated in selective growth inhibition and apoptosis in neoplastic cells. On the basis of mechanistic studies and other recent developments, we propose that FTIs may be useful to treat a unique spectrum of diseases including not only inflammatory breast cancer and melanoma but also non-neoplastic diseases such as diabetic retinopathy and macular degeneration.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Alkyl and Aryl Transferases/genetics , Animals , Clinical Trials as Topic , Farnesyltranstransferase , Humans , Neoplasms/enzymology , Neoplasms/pathology , rhoB GTP-Binding Protein/antagonists & inhibitors
16.
Oncogene ; 19(41): 4669-84, 2000 Sep 28.
Article in English | MEDLINE | ID: mdl-11032017

ABSTRACT

Cell death processes are progressively inactivated during malignant development, in part by loss of tumor suppressors that can promote cell death. The Bin1 gene encodes a nucleocytosolic adaptor protein with tumor suppressor properties, initially identified through its ability to interact with and inhibit malignant transformation by c-Myc and other oncogenes. Bin1 is frequently missing or functionally inactivated in breast and prostate cancers and in melanoma. In this study, we show that Bin1 engages a caspase-independent cell death process similar to type II apoptosis, characterized by cell shrinkage, substratum detachment, vacuolated cytoplasm, and DNA degradation. Cell death induction was relieved by mutation of the BAR domain, a putative effector domain, or by a missplicing event that occurs in melanoma and inactivates suppressor activity. Cells in all phases of the cell cycle were susceptible to death and p53 and Rb were dispensable. Notably, Bin1 did not activate caspases and the broad spectrum caspase inhibitor ZVAD.fmk did not block cell death. Consistent with the lack of caspase involvement, dying cells lacked nucleosomal DNA cleavage and nuclear lamina degradation. Moreover, neither Bcl-2 or dominant inhibition of the Fas pathway had any effect. In previous work, we showed that Bin1 could not suppress cell transformation by SV40 large T antigen. Consistent with this finding, we observed that T antigen suppressed the death program engaged by Bin1. This observation was interesting in light of emerging evidence that T antigen has roles in cell immortalization and human cell transformation beyond Rb and p53 inactivation. In support of a link to c-Myc-induced death processes, AEBSF, a serine protease inhibitor that inhibits apoptosis by c-Myc, potently suppressed DNA degradation by Bin1. Our findings suggest that the tumor suppressor activity of Bin1 reflects engagement of a unique cell death program. We propose that loss of Bin1 may promote malignancy by blunting death penalties associated with oncogene activation.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Caspases/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Chloromethyl Ketones/pharmacology , Antigens, Polyomavirus Transforming/physiology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Adhesion , Cell Size , Cell Transformation, Neoplastic/genetics , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mitochondria/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/physiology , Retinoblastoma Protein/physiology , Serine Proteinase Inhibitors/pharmacology , Sulfones/pharmacology , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/physiology , fas Receptor/physiology
17.
FEBS Lett ; 481(3): 205-8, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-11007964

ABSTRACT

Farnesyltransferase inhibitors (FTIs) induce apoptosis by elevating the levels of geranylgeranylated RhoB (RhoB-GG) in cells. However, the mechanism by which RhoB-GG acts is unclear. Here we report that RhoB-GG is sufficient to mediate the suppressive effects of FTIs on the activity of the survival kinase Akt-1 in epithelial cells. This mechanism is tissue-specific insofar as it does not operate in fibroblasts. We discuss how the cell survival functions of RhoB and Akt may be linked biochemically in certain cell types.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Protein Prenylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Retroviridae Proteins, Oncogenic/antagonists & inhibitors , rhoB GTP-Binding Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , COS Cells , Enzyme Activation/drug effects , Enzyme Repression/drug effects , Epidermal Growth Factor/pharmacology , Farnesyltranstransferase , Genes, ras , Humans , Oncogene Protein v-akt , Organ Specificity , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/physiology , Retroviridae Proteins, Oncogenic/biosynthesis , Retroviridae Proteins, Oncogenic/metabolism , Tumor Cells, Cultured , rhoB GTP-Binding Protein/physiology
18.
Mol Cell Biol ; 20(16): 6105-13, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913192

ABSTRACT

Farnesyltransferase inhibitors (FTIs) are in clinical trials, but how they selectively inhibit malignant cell growth remains uncertain. One important player in this process appears to be RhoB, an endosomal Rho protein that regulates receptor trafficking. FTI treatment elicits a gain of the geranylgeranylated RhoB isoform (RhoB-GG) that occurs due to modification of RhoB by geranylgeranyltransferase I in drug-treated cells. Notably, this event is sufficient to mediate antineoplastic effects in murine models and human carcinoma cells. To further assess this gain-of-function mechanism and determine whether RhoB-GG has a necessary role in drug action, we examined the FTI response of murine fibroblasts that cannot express RhoB-GG due to homozygous deletion of the rhoB gene. Nullizygous (-/-) cells were susceptible to cotransformation by adenovirus E1A plus activated H-Ras but defective in their FTI response, despite complete inhibition of H-Ras prenylation. Actin cytoskeletal and phenotypic events were disrupted in -/- cells, implicating RhoB-GG in these effects. Interestingly, -/- cells were resistant to FTI-induced growth inhibition under anchorage-dependent but not anchorage-independent conditions, indicating that, while RhoB-GG is sufficient, it is not necessary for growth inhibition under all conditions. In contrast, -/- cells were resistant to FTI-induced apoptosis in vitro and in vivo. Significantly, the apoptotic defect of -/- cells compromised the antitumor efficacy of FTI in xenograft assays. This study offers genetic proof of the hypothesis that RhoB-GG is a crucial mediator of the antineoplastic effects of FTIs.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Enzyme Inhibitors/pharmacology , rhoB GTP-Binding Protein/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Farnesyltranstransferase , Humans , Mice
19.
Genomics ; 67(2): 210-20, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10903846

ABSTRACT

BAR family proteins are a unique class of adaptor proteins characterized by a common N-terminal fold of undetermined function termed the BAR domain. This set of adaptors, which includes the mammalian proteins amphiphysin and Bin1 and the yeast proteins Rvs167p and Rvs161p, has been implicated in diverse cellular processes, including synaptic vesicle endocytosis, actin regulation, differentiation, cell survival, and tumorigenesis. Here we report the identification and characterization of Bin2, a novel protein that contains a BAR domain but that is otherwise structurally dissimilar to other members of the BAR adaptor family. The Bin2 gene is located at chromosome 4q22.1 and is expressed predominantly in hematopoietic cells. Bin2 is upregulated during differentiation of granulocytes, suggesting that it functions in that lineage. Bin2 formed a stable complex in cells with Bin1, but not with amphiphysin, in a BAR domain-dependent manner. This finding indicates that BAR domains have specific preferences for interaction. However, Bin2 did not influence endocytosis in the same manner as brain-specific splice isoforms of Bin1, nor did it exhibit the tumor suppressor properties inherent to ubiquitous splice isoforms of Bin1. Thus, Bin2 appears to encode a nonredundant function in the BAR adaptor gene family.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Chromosomes, Human, Pair 4/genetics , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Carrier Proteins/metabolism , Cell Line , Chromosome Banding , Chromosome Mapping , Conserved Sequence , Cytosol/chemistry , Cytosol/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , Endocytosis , Female , Gene Expression , Genes, Tumor Suppressor , HL-60 Cells , Humans , In Situ Hybridization, Fluorescence , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA/genetics , RNA/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured , src Homology Domains
20.
Int J Cancer ; 86(2): 155-61, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10738240

ABSTRACT

The genetic events underlying the development of prostate cancer are poorly defined. c-Myc is often activated in tumors that have progressed to metastatic status, so events that promote this process may be important. Bin1 is a nucleocytoplasmic adaptor protein with features of a tumor suppressor that was identified through its ability to interact with and inhibit malignant transformation by c-Myc. We investigated a role for Bin1 loss or inactivation in prostate cancer because the human Bin1 gene is located at chromosome 2q14 within a region that is frequently deleted in metastatic prostate cancer but where no tumor suppressor candidate has been located. A novel polymorphic microsatellite marker located within intron 5 of the human Bin1 gene was used to demonstrate loss of heterozygosity and coding alteration in 40% of informative cases of prostate neoplasia examined. RNA and immunohistochemical analyses indicated that Bin1 was expressed in most primary tumors, even at slightly elevated levels relative to benign tissues, but that it was frequently missing or inactivated by aberrant splicing in metastatic tumors and androgen-independent tumor cell lines. Ectopic expression of Bin1 suppressed the growth of prostate cancer lines in vitro. Our findings support the candidacy of Bin1 as the chromosome 2q prostate tumor suppressor gene.


Subject(s)
Carrier Proteins/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Adenocarcinoma/genetics , Alternative Splicing , Blotting, Northern , Genes, myc , Humans , Immunohistochemistry , Introns , Male , Microsatellite Repeats , Prostatic Intraepithelial Neoplasia/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL