Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncotarget ; 5(15): 6252-66, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25051369

ABSTRACT

Velcade is one of the inescapable drug to treat patient suffering from multiple myeloma (MM) and resistance to this drug represents a major drawback for patients. However, the mechanisms underlying velcade resistance remain incompletely understood. We derived several U266 MM cell clones that resist to velcade. U266-resistant cells were resistant to velcade-induced cell death but exhibited a similar sensitivity to various proapoptotic stimuli. Careful analysis of proteosomal subunits and proteasome enzymatic activities showed that neither the composition nor the activity of the proteasome was affected in velcade-resistant cells. Elimination of velcade-induced poly-ubiquitinated proteins and protein aggregates was drastically stimulated in the resistant cells and correlated with increased cell survival. Inhibition of the lysosomal activity in velcade-resistant cells resulted in an increase of cell aggregates and decrease survival, indicating that aggregates are eliminated through lysosomal degradation. In addition, pangenomic profiling of velcade-sensitive and resistant cells showed that the small heat shock protein HSPB8 was overexpressed in resistant cells. Finally, gain and loss of function experiment demonstrated that HSPB8 is a key factor for velcade resistance. In conclusion, HSPB8 plays an important role for the elimination of aggregates in velcade-resistant cells that contributes to their enhanced survival.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Heat-Shock Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Molecular Chaperones , Multiple Myeloma/pathology , Protein Folding
2.
PLoS One ; 7(10): e47321, 2012.
Article in English | MEDLINE | ID: mdl-23071785

ABSTRACT

Transgenic mice expressing the caspase-cleaved form of the tyrosine kinase Lyn (LynΔN) develop a TNFα-dependent skin disease that accurately recapitulates human psoriasis. Participation of lymphocytes in this disease was confirmed by backcrossing LynΔN mice on a Rag-1 deficient background. The present study was therefore conducted to analyze whether modification of lymphocyte homeostasis does occur and participate in the phenotype of LynΔN mice. We show here that LynΔN mice consistently exhibit thymic atrophy that correlates with both a net decrease in the CD4+/CD8+ Double Positive (DP) and an increase in Single Positive (SP) thymocyte sub-populations, but also display an increase of splenic mature B cell. Interestingly, a normal immune phenotype was rescued in a TNFR1 deficient background. Finally, none of these immune alterations was detected in newborn mice before the onset of inflammation. Therefore, we conclude that chronic inflammation can induce thymic atrophy and perturb spleen homeostasis in LynΔN mice through the increased production of TNFα, LTß and TNFR1 signaling.


Subject(s)
Dermatitis/physiopathology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , Thymus Gland/pathology , src-Family Kinases/genetics , Animals , Atrophy/physiopathology , B-Lymphocytes/cytology , Bromodeoxyuridine , Dermatitis/genetics , Flow Cytometry , Immunoblotting , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Tumor Necrosis Factor-alpha/metabolism
3.
Autophagy ; 8(4): 637-49, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22498477

ABSTRACT

Bcl-2 family members are key modulators of apoptosis that have recently been shown to also regulate autophagy. It has been previously reported that Bcl-2 and Bcl-X(L) bind and inhibit BECN1, an essential mediator of autophagy. Bcl-B is an anti-apoptotic member of the Bcl-2 family that possesses the four BH (Bcl-2 homology) domains (BH1, BH2, BH3 and BH4) and a predicted C-terminal trans-membrane domain. Although the anti-apoptotic properties of Bcl-B are well characterized, its physiological function remains to be established. In the present study, we first established that Bcl-B interacts with the BH3 domain of BECN1. We also showed that Bcl-B overexpression reduces autophagy triggered by a variety of pro-autophagic stimuli. This impairment of autophagy was closely related to the capacity of Bcl-B to bind to BECN1. Importantly, we have demonstrated that Bcl-B knockdown triggers autophagic cell death and sensitizes cells to amino acid starvation. The cell death induced by Bcl-B knockdown was partially dependent on components of the autophagy machinery (LC3; BECN1; ATG5). These findings reveal a new role of Bcl-B in the regulation of autophagy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Autophagy , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Amino Acids/deficiency , Beclin-1 , Gene Knockdown Techniques , Gene Silencing , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry
4.
Oncotarget ; 2(11): 874-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22141136

ABSTRACT

AXL is a receptor tyrosine kinase of the TAM family, the function of which is poorly understood. We previously identified AXL overexpression in Imatinib (IM)-resistant CML cell lines and patients. The present study was conducted to investigate the role of AXL and the mechanisms underlying AXL overexpression in Tyrosine Kinase Inhibitor (TKI)-resistant CML cells. We present evidence that high AXL expression level is a feature of TKI-resistant CML cells and knockdown of AXL sensitized TKI-resistant cells to IM. In addition, expression of wild-type AXL but not a dominant negative form of AXL confers IM-sensitive CML cells the capacity to resist IM effect. AXL overexpression required PKCα and ß and constitutive activation of ERK1/2. Accordingly, GF109203X a PKC inhibitor, U0126 a MEK1 inhibitor and PKCα/ß knockdown restore sensitivity to IM while PKCα or PKCß overexpression in CML cells promotes protection against IM-induced cell death. Finally, using luciferase promoter activity assays we established that AXL is regulated transcriptionally through the AP1 transcription factor. Our findings reveal an unexpected role of AXL in resistance to TKI in CML cells, identify the molecular mechanisms involved in its overexpression and support the notion that AXL is a new marker of resistance to TKI in CML.


Subject(s)
Drug Resistance, Neoplasm/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/physiology , Transfection , Up-Regulation , Axl Receptor Tyrosine Kinase
5.
PLoS One ; 4(11): e7889, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19924252

ABSTRACT

CML is an hematopoietic stem cell disease characterized by the t(9;22) (q34;q11) translocation encoding the oncoprotein p210BCR-ABL. The effect of acadesine (AICAR, 5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside) a compound with known antileukemic effect on B cell chronic lymphoblastic leukemia (B-CLL) was investigated in different CML cell lines. Acadesine triggered loss of cell metabolism in K562, LAMA-84 and JURL-MK1 and was also effective in killing imatinib-resistant K562 cells and Ba/F3 cells carrying the T315I-BCR-ABL mutation. The anti-leukemic effect of acadesine did not involve apoptosis but required rather induction of autophagic cell death. AMPK knock-down by Sh-RNA failed to prevent the effect of acadesine, indicating an AMPK-independent mechanism. The effect of acadesine was abrogated by GF109203X and Ro-32-0432, both inhibitor of classical and new PKCs and accordingly, acadesine triggered relocation and activation of several PKC isoforms in K562 cells. In addition, this compound exhibited a potent anti-leukemic effect in clonogenic assays of CML cells in methyl cellulose and in a xenograft model of K562 cells in nude mice. In conclusion, our work identifies an original and unexpected mechanism by which acadesine triggers autophagic cell death through PKC activation. Therefore, in addition to its promising effects in B-CLL, acadesine might also be beneficial for Imatinib-resistant CML patients.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Autophagy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , K562 Cells , Mice , Mice, Nude , Protein Kinase C/metabolism , RNA, Small Interfering/metabolism , Translocation, Genetic
6.
EMBO J ; 28(16): 2449-60, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19590497

ABSTRACT

We showed previously that Lyn is a substrate for caspases, a family of cysteine proteases, involved in the regulation of apoptosis and inflammation. Here, we report that expression of the caspase-cleaved form of Lyn (LynDeltaN), in mice, mediates a chronic inflammatory syndrome resembling human psoriasis. Genetic ablation of TNF receptor 1 in a LynDeltaN background rescues a normal phenotype, indicating that LynDeltaN mice phenotype is TNF-alpha-dependent. The predominant role of T cells in the disease occurring in LynDeltaN mice was highlighted by the distinct improvement of LynDeltaN mice phenotype in a Rag1-deficient background. Using pan-genomic profiling, we also established that LynDeltaN mice show an increased expression of STAT-3 and inhibitory members of the NFkappaB pathway. Accordingly, LynDeltaN alters NFkappaB activity underlying a link between inhibition of NFkappaB and LynDeltaN mice phenotype. Finally, analysis of Lyn expression in human skin biopsies of psoriatic patients led to the detection of Lyn cleavage product whose expression correlates with the activation of caspase 1. Our data identify a new role for Lyn as a regulator of psoriasis through its cleavage by caspases.


Subject(s)
Psoriasis/metabolism , Skin/pathology , src-Family Kinases/genetics , src-Family Kinases/metabolism , Animals , Biopsy , Caspases/metabolism , Cells, Cultured , Gene Deletion , Gene Expression , Humans , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phenotype , Psoriasis/genetics , Skin/anatomy & histology , Thymus Gland/cytology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
FASEB J ; 22(6): 1894-904, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18245170

ABSTRACT

Imatinib is successfully used in the treatment of chronic myelogenous leukemia (CML), and the main mechanisms of resistance in refractory patients are now partially understood. In the present study, we investigated the mechanism of action of resveratrol in imatinib-sensitive (IM-S) and -resistant (IM-R) CML cell lines. Resveratrol induced loss of viability and apoptosis in IM-S and IM-R in a time- and dose-dependent fashion. Inhibition of cell viability was detected for concentrations of resveratrol as low as 5 microM, and the IC(50) values for viability, clonogenic assays, apoptosis, and erythroid differentiation were in the 10-25 microM range. The effect of imatinib and resveratrol was additive in IM-S but not in IM-R clones in which the resveratrol effect was already maximal. The effect of resveratrol on apoptosis was partially rescued by zVAD-fmk, suggesting a caspase-independent contribution. Resveratrol action was independent of BCR-ABL expression and phosphorylation, and in agreement was additive to BCR-ABL silencing. Finally, phytoalexin inhibited the growth of BaF3 cells expressing mutant BCR-ABL proteins found in resistant patients, including the multiresistant T315I mutation. Our findings show that resveratrol induces apoptosis, caspase-independent death, and differentiation that collectively contribute to the specific elimination of CML cells. Resveratrol should provide therapeutic benefits in IM-R patients and in other hematopoietic malignancies.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic , Apoptosis/drug effects , Benzamides , Caspases , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Resveratrol
8.
J Invest Surg ; 20(2): 97-103, 2007.
Article in English | MEDLINE | ID: mdl-17454394

ABSTRACT

In hemorrhagic shock (HS) an increased thymic apoptosis (TA) was described. The aim of this study was to evaluate the effect of administration of the caspase inhibitor N-benzyloxy-carbonil-Val-Ala-Asp-fluoromethylketone (Z-VAD-FMK) during the resuscitation phase on TA, organ dysfunctions, and tumor necrosis factor (TNF)-alpha release in HS. Forty rats were randomly assigned to four groups: no HS/resuscitation (sham); HS/resuscitation with shed blood and normal saline (control); HS/resuscitation with shed blood and phosphate-buffered solution (PBS) (vehicle); and HS/resuscitation with shed blood and Z-VAD-FMK (inhibitor). Rats were subjected to HS by blood removal to a MAP of 35-40 mmHg. After a 1-h shock period, the animals were resuscitated according to the protocol. At 1 and 3 h after resuscitation, transaminases, creatinine, urea, lipase, TNF-alpha, and TA were evaluated. Our study showed that a nonlethal HS is early able to induce organ dysfunctions and increased TA. Administration of Z-VAD-FMK did not significantly decrease organ dysfunctions, while it induced a significant TNF-alpha release. TA was significantly reduced by Z-VAD-FMK after 1 h, but not after 3 h. Our results suggest that postinjury caspase inhibition does not attenuate organ dysfunctions, and also does not permanently reduce TA induced by HS and resuscitation in rats.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Shock, Hemorrhagic/pathology , Thymus Gland/drug effects , Thymus Gland/pathology , Animals , Caspases/metabolism , Male , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/metabolism
9.
Oncogene ; 23(47): 7863-73, 2004 Oct 14.
Article in English | MEDLINE | ID: mdl-15378010

ABSTRACT

Mcl-1 is an antiapoptotic member of the Bcl-2 family that can promote cell viability. We report here that Mcl-1 is a new substrate for caspases during induction of apoptosis. Mcl-1 cleavage occurs after Asp127 and Asp157 and generates four fragments of 24, 19, 17 and 12 kDa in both intact cells and in vitro, an effect prevented by selective caspase inhibitors. As a consequence, the resulting protein that lacks the first 127 or 157 amino acids contains only the BH1-BH3 domains of Bcl-2 family members. Mutation of Asp127 and Asp157 abolishes the generation of the 24 and 12 kDa fragments and that of the 19 and 17 kDa fragments, respectively. Interestingly, when expressed in HeLa cells Mcl-1 wt and Mcl-1 Delta127 showed a markedly different intracellular distribution. Mcl-1 wt colocalized with alpha-Tubulin near the internal face of the plasma membrane, while Mcl-1 Delta127 coassociated with Bim-EL at the mitochondrial level. Coimmunoprecipitation experiments also demonstrated that Mcl1 Delta127 exhibited increased binding to Bim when compared to Mcl-1 wt. Finally, Mcl-1 wt unlike Mcl-1 Delta127 inhibited Bim-EL-induced caspase activation. Altogether, our findings demonstrate that cleavage of Mcl-1 by caspases modifies its subcellular localization, increases its association with Bim and inhibits its antiapoptotic function.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Caspases/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/metabolism , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Carrier Proteins/genetics , DNA Primers , Flow Cytometry , HeLa Cells , Humans , Jurkat Cells , K562 Cells , Membrane Proteins/genetics , Microscopy, Confocal , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Protein Biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Transcription, Genetic , Transfection
11.
J Bone Miner Res ; 18(10): 1863-71, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14584897

ABSTRACT

UNLABELLED: Molecular events of mechanotransduction in osteoblasts are poorly defined. We show that the mechanosensitive BK channels open and recruit the focal adhesion kinase FAK in osteoblasts on hypotonic shock. This could convert mechanical signals in biochemical events, leading to osteoblast activation. INTRODUCTION: Mechanical strains applied to the skeleton influence bone remodeling and architecture mainly through the osteoblast lineage. The molecular mechanisms involved in osteoblastic mechanotransduction include opening of mechanosensitive cation channels and the activation of protein tyrosine kinases, notably FAK, but their interplay remains poorly characterized. The large conductance K+ channel (BK) seems likely as a bone mechanoreceptor candidate because of its high expression in osteoblasts and its ability to open in response to membrane stretch or hypotonic shock. Propagation of the signals issued from the mechanosensitivity of BK channels inside the cell likely implies complex interactions with molecular partners involved in mechanotransduction, notably FAK. METHODS: Interaction of FAK with the C terminus of the hSlo alpha-subunit of BK was investigated using the yeast two-hybrid system as well as immunofluorescence microscopy and coimmunoprecipitation experiments with a rabbit anti-hslo antibody on MG63 and CAL72 human osteosarcoma cell lines and on normal human osteoblasts. Mapping of the FAK region interacting with hSlo was approached by testing the ability of hSlo to recruit mutated ot truncated FAK proteins. RESULTS: To the best of our knowledge, we provide the first evidence of the physical association of FAK with the intracellular part of hslo. We show that FAK/hSlo interaction likely takes place through the Pro-1-rich domain situated in the C-terminal region of the kinase. FAK/hSlo association occurs constitutively at a low, but appreciable, level in human osteosarcoma cells and normal human osteoblasts that express endogenous FAK and hSlo. In addition, we found that application of an hypo-osmotic shock to these cells induced a sustained activation of BK channels associated to a marked increase in the recruitment of FAK on hSlo. CONCLUSIONS: Based on these data, we propose that BK channels might play a triggering role in the signaling cascade induced by mechanical strains in osteoblasts.


Subject(s)
Calcium/metabolism , Osteoblasts/metabolism , Potassium Channels, Calcium-Activated/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , COS Cells , Cell Line , Cell Lineage , Electrophysiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Osteoblasts/pathology , Plasmids/metabolism , Potassium Channels/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Two-Hybrid System Techniques
12.
Int J Cancer ; 106(3): 355-63, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12845673

ABSTRACT

Stromelysin-3 (ST3) has the characteristic structure of matrix metalloproteinases (MMP), but its substrate specificity and pattern of expression differ markedly from that of other MMP family members. ST3 was originally isolated on the basis of its expression in primary breast cancers and has been shown to be overexpressed in virtually all primary carcinomas, suggesting that ST3 participates in the initial development of epithelial malignancies. Recent data using murine models reported that ST3 expression was able to increase tumor take by suppressing cell apoptosis. Our present goal was to set up an in vitro model in which we could study this new function. For this purpose, we analyzed survival of MCF-7 transfectants expressing either wild-type or catalytically inactive ST3 (ST3wt or ST3cat-) in three-dimensional (3-D) culture conditions by inclusion in Matrigel. In such conditions, that mimic the in vivo microenvironment, we found a marked decrease in the percentage of cell death when active ST3 was expressed (ST3wt transfectants vs. ST3cat- or vector only transfectants) as assessed by FACS and TUNEL analysis. The addition of batimastat, a broad spectrum MMP inhibitor, reversed the increased cell survival in ST3wt transfectants, confirming that ST3 enzymatic activity was required for this effect. Finally, we analyzed the expression of anti- and pro-apoptotic proteins as well as activation of cell survival pathways and we found that ST3-mediated cell survival was accompanied by activation of both p42/p44 MAPK and AKT. Our data confirm and extend the anti-apoptotic function of ST3 and provide a useful model to dissect this new role and identify new physiological substrates.


Subject(s)
Breast Neoplasms/enzymology , Metalloendopeptidases/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phenylalanine/analogs & derivatives , Protein Serine-Threonine Kinases , Antineoplastic Agents/pharmacology , Blotting, Western , Breast Neoplasms/pathology , Caspase Inhibitors , Cell Survival/physiology , Collagen , Drug Combinations , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Humans , In Situ Nick-End Labeling , Laminin , Matrix Metalloproteinase 11 , Metalloendopeptidases/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mutagenesis, Site-Directed , Mutation , Phenylalanine/pharmacology , Phosphorylation , Proteoglycans , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Thiophenes/pharmacology , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...