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










Database
Publication year range
1.
J Biol Chem ; 287(26): 21796-805, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22556409

ABSTRACT

Activation of PI3-K-AKT and ERK pathways is a complication of mTOR inhibitor therapy. Newer mTOR inhibitors (like pp242) can overcome feedback activation of AKT in multiple myeloma (MM) cells. We, thus, studied if feedback activation of ERK is still a complication of therapy with such drugs in this tumor model. PP242 induced ERK activation in MM cell lines as well as primary cells. Surprisingly, equimolar concentrations of rapamycin were relatively ineffective at ERK activation. Activation was not correlated with P70S6kinase inhibition nor was it prevented by PI3-kinase inhibition. ERK activation was prevented by MEK inhibitors and was associated with concurrent stimulation of RAF kinase activity but not RAS activation. RAF activation correlated with decreased phosphorylation of RAF at Ser-289, Ser-296, and Ser-301 inhibitory residues. Knockdown studies confirmed TORC1 inhibition was the key proximal event that resulted in ERK activation. Furthermore, ectopic expression of eIF-4E blunted pp242-induced ERK phosphorylation. Since pp242 was more potent than rapamycin in causing sequestering of eIF-4E, a TORC1/4E-BP1/eIF-4E-mediated mechanism of ERK activation could explain the greater effectiveness of pp242. Use of MEK inhibitors confirmed ERK activation served as a mechanism of resistance to the lethal effects of pp242. Thus, although active site mTOR inhibitors overcome AKT activation often seen with rapalog therapy, feedback ERK activation is still a problem of resistance, is more severe than that seen with use of first generation rapalogs and is mediated by a TORC1- and eIF-4E-dependent mechanism ultimately signaling to RAF.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Indoles/pharmacology , Multiple Myeloma/metabolism , Purines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Apoptosis , Catalytic Domain , Cell Line, Tumor , Cell Survival , Enzyme Activation , Glutathione Transferase/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiple Myeloma/drug therapy , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism , Signal Transduction , Sirolimus/pharmacology
2.
J Biol Chem ; 286(1): 67-78, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-20974848

ABSTRACT

Prior work indicates that IL-6 can stimulate c-Myc expression in multiple myeloma (MM) cells, which is independent of effects on transcription and due to enhanced translation mediated by an internal ribosome entry site in the 5'-UTR of the c-Myc RNA. The RNA-binding protein hnRNP A1 (A1) was also critical to IL-6-stimulated translation. Because A1 shuttles between nucleus and cytoplasm, we investigated whether the ability of IL-6 to enhance Myc translation was mediated by stimulation of A1 shuttling. In MM cell lines and primary specimens, IL-6 increased A1 cytoplasmic localization. In contrast, there was no effect on the total cellular levels of A1. Use of a dominant negative A1 construct, which prevents endogenous A1 from nucleus-to-cytoplasm transit, prevented the ability of IL-6 to enhance Myc internal ribosome entry site activity, Myc protein expression, and MM cell growth. IL-6-stimulated cytoplasmic localization was mediated by alterations in the C-terminal M9 peptide of A1, and this correlated with the ability of IL-6 to induce serine phosphorylation of this domain. A p38 kinase inhibitor prevented IL-6-induced A1 phosphorylation. Thus, IL-6 activates c-Myc translation in MM cells by inducing A1 phosphorylation and cytoplasmic localization in a p38-dependent fashion. These data suggest A1 as a potential therapeutic target in MM.


Subject(s)
Cytoplasm/drug effects , Cytoplasm/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Interleukin-6/pharmacology , Multiple Myeloma/pathology , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-myc/biosynthesis , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Mutation , Peptide Fragments/metabolism , Phosphorylation/drug effects
3.
Blood ; 116(22): 4560-8, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20686120

ABSTRACT

Although preclinical work with rapalogs suggests potential in treatment of multiple myeloma (MM), they have been less successful clinically. These drugs allostearically inhibit the mammalian target of rapamycin kinase primarily curtailing activity of the target of rapamycin complex (TORC)1. To assess if the mammalian target of rapamycin within the TORC2 complex could be a better target in MM, we tested a new agent, pp242, which prevents activation of TORC2 as well as TORC1. Although comparable to rapamycin against phosphorylation of the TORC1 substrates p70S6kinase and 4E-BP-1, pp242 could also inhibit phosphorylation of AKT on serine 473, a TORC2 substrate, while rapamycin was ineffective. pp242 was also more effective than rapamycin in achieving cytoreduction and apoptosis in MM cells. In addition, pp242 was an effective agent against primary MM cells in vitro and growth of 8226 cells in mice. Knockdown of the TORC2 complex protein, rictor, was deleterious to MM cells further supporting TORC2 as the critical target for pp242. TORC2 activation was frequently identified in primary specimens by immunostaining for AKT phosphorylation on serine 473. Potential mechanisms of up-regulated TORC2 activity in MM were stimulation with interleukin-6 or insulin-like growth factor 1, and phosphatase and tensin homolog or RAS alterations. Combining pp242 with bortezomib led to synergistic anti-MM effects. These results support TORC2 as a therapeutic target in MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Carrier Proteins/genetics , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Drug Synergism , Gene Knockdown Techniques , Humans , Mice , Mice, SCID , Multiprotein Complexes/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Rapamycin-Insensitive Companion of mTOR Protein
4.
Mol Cancer Ther ; 8(7): 1974-84, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19509276

ABSTRACT

Because accumulation of potentially toxic malfolded protein may be extensive in immunoglobulin-producing multiple myeloma (MM) cells, we investigated the phenomenon of autophagy in myeloma, a physiologic process that can protect against malfolded protein under some circumstances. Autophagy in MM cell lines that express and secrete immunoglobulin and primary specimens was significantly increased by treatment with the endoplasmic reticulum stress-inducing agent thapsigargin, the mammalian target of rapamycin inhibitor rapamycin, and the proteasome inhibitor bortezomib. Inhibition of basal autophagy in these cell lines and primary cells by use of the inhibitors 3-methyladenine and chloroquine resulted in a cytotoxic effect that was associated with enhanced apoptosis. Use of small interfering RNA to knock down expression of beclin-1, a key protein required for autophagy, also inhibited viable recovery of MM cells. Because the data suggested that autophagy protected MM cell viability, we predicted that autophagy inhibitors would synergize with bortezomib for enhanced antimyeloma effects. However, the combination of these drugs resulted in an antagonistic response. In contrast, the autophagy inhibitor 3-methyladenine did synergize with thapsigargin for an enhanced cytotoxic response. These data suggest that autophagy inhibitors have therapeutic potential in myeloma but caution against combining such drugs with bortezomib.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Multiple Myeloma/pathology , Adenine/analogs & derivatives , Adenine/pharmacology , Antifungal Agents/pharmacology , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Boronic Acids/pharmacology , Bortezomib , Cell Proliferation/drug effects , Chloroquine/pharmacology , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Humans , Immunoblotting , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Fluorescence , Multiple Myeloma/drug therapy , Pyrazines/pharmacology , RNA, Small Interfering/pharmacology , Sirolimus/pharmacology , Thapsigargin/pharmacology , Tumor Cells, Cultured
5.
Cancer Res ; 68(24): 10215-22, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19074889

ABSTRACT

Prior work indicates that c-myc translation is up-regulated in multiple myeloma cells. To test a role for interleukin (IL)-6 in myc translation, we studied the IL-6-responsive ANBL-6 and IL-6-autocrine U266 cell lines as well as primary patient samples. IL-6 increased c-myc translation, which was resistant to rapamycin, indicating a mechanism independent of mammalian target of rapamycin (mTOR) and cap-dependent translation. In contrast, the cytokine enhanced cap-independent translation via a stimulatory effect on the myc internal ribosome entry site (IRES). As known IRES-trans-activating factors (ITAF) were unaffected by IL-6, we used a yeast-three-hybrid screen to identify novel ITAFs and identified hnRNP A1 (A1) as a mediator of the IL-6 effect. A1 specifically interacted with the myc IRES in filter binding assays as well as EMSAs. Treatment of myeloma cells with IL-6 induced serine phosphorylation of A1 and increased its binding to the myc IRES in vivo in myeloma cells. Primary patient samples also showed binding between A1 and the IRES. RNA interference to knock down hnRNP A1 prevented an IL-6 increase in myc protein expression, myc IRES activity, and cell growth. These data point to hnRNP A1 as a critical regulator of c-myc translation and a potential therapeutic target in multiple myeloma.


Subject(s)
Genes, myc , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Interleukin-6/pharmacology , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Ribosomes/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-myc/genetics , Ribosomes/genetics , Transfection
6.
La Paz; 2003. 69 p. ilus.
Thesis in Spanish | LIBOCS, LIBOSP | ID: biblio-1312413

ABSTRACT

El objetivo principal de este proyecto de grado es efectuar una comparación de la operación de la línea Expreso Villa Adela-Plaza Alonzo de Mendoza, con relación al sistema convencional de minibuses y micros, que sisrven como transporte público en La ciudad de La Paz.

SELECTION OF CITATIONS
SEARCH DETAIL
...