Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Oncogene ; 39(27): 5001-5014, 2020 07.
Article in English | MEDLINE | ID: mdl-32523091

ABSTRACT

Proteasome inhibitors have provided a significant advance in the treatment of multiple myeloma (MM). Consequently, there is increasing interest in developing strategies to target E3 ligases, de-ubiquitinases, and/or ubiquitin receptors within the ubiquitin proteasome pathway, with an aim to achieve more specificity and reduced side-effects. Previous studies have shown a role for the E3 ligase HUWE1 in modulating c-MYC, an oncogene frequently dysregulated in MM. Here we investigated HUWE1 in MM. We identified elevated expression of HUWE1 in MM compared with normal cells. Small molecule-mediated inhibition of HUWE1 resulted in growth arrest of MM cell lines without significantly effecting the growth of normal bone marrow cells, suggesting a favorable therapeutic index. Studies using a HUWE1 knockdown model showed similar growth inhibition. HUWE1 expression positively correlated with MYC expression in MM bone marrow cells and correspondingly, genetic knockdown and biochemical inhibition of HUWE1 reduced MYC expression in MM cell lines. Proteomic identification of HUWE1 substrates revealed a strong association of HUWE1 with metabolic processes in MM cells. Intracellular glutamine levels are decreased in the absence of HUWE1 and may contribute to MYC degradation. Finally, HUWE1 depletion in combination with lenalidomide resulted in synergistic anti-MM activity in both in vitro and in vivo models. Taken together, our data demonstrate an important role of HUWE1 in MM cell growth and provides preclinical rationale for therapeutic strategies targeting HUWE1 in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Lenalidomide/pharmacology , Multiple Myeloma/drug therapy , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Bone Marrow Cells/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , RNA Interference , RNA, Small Interfering/genetics , Therapeutic Index, Drug , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
2.
J Cell Commun Signal ; 12(1): 21-29, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29110249

ABSTRACT

Post-translational modification of proteins with ubiquitin plays a central role in regulating numerous cellular processes. E3 ligases determine the specificity of ubiquitination by mediating the transfer of ubiquitin to substrate proteins. The family of tripartite motif (TRIM) proteins make up one of the largest subfamilies of E3 ligases. Accumulating evidence suggests that dysregulation of TRIM proteins is associated with a variety of diseases. In this review we focus on the involvement of TRIM proteins in blood cancers.

3.
Methods Mol Biol ; 1489: 261-271, 2017.
Article in English | MEDLINE | ID: mdl-27734383

ABSTRACT

The regulation of blood cell production (hematopoiesis) by CCN proteins is an area of increasing interest to hematologists. There is some discordance in the literature in this area due to the use of mixed or ill-defined cell populations for experiments. Expression of, and response to, CCN proteins is specific to both cell type and differentiation status. Here, we describe methods to prepare defined hematopoietic cell populations and associated functional assays.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Cell Separation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Biomarkers , CCN Intercellular Signaling Proteins/genetics , Cell Line , Cell Separation/methods , Coculture Techniques , Colony-Forming Units Assay , Feeder Cells , Flow Cytometry , Gene Expression , Hematopoiesis , Humans , Immunomagnetic Separation/methods
4.
Oncotarget ; 7(43): 70481-70493, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27655696

ABSTRACT

Multiple Myeloma (MM) is a haematological neoplasm characterised by the clonal proliferation of malignant plasma cells in the bone marrow. The success of proteasome inhibitors in the treatment of MM has highlighted the importance of the ubiquitin proteasome system (UPS) in the pathogenesis of this disease. In this study, we analysed gene expression of UPS components to identify novel therapeutic targets within this pathway in MM. Here we demonstrate how this approach identified previously validated and novel therapeutic targets. In addition we show that FZR1 (Fzr), a cofactor of the multi-subunit E3 ligase complex anaphase-promoting complex/cyclosome (APC/C), represents a novel therapeutic target in myeloma. The APC/C associates independently with two cofactors, Fzr and Cdc20, to control cell cycle progression. We found high levels of FZR1 in MM primary cells and cell lines and demonstrate that expression is further increased on adhesion to bone marrow stromal cells (BMSCs). Specific knockdown of either FZR1 or CDC20 reduced viability and induced growth arrest of MM cell lines, and resulted in accumulation of APC/CFzr substrate Topoisomerase IIα (TOPIIα) or APC/CCdc20 substrate Cyclin B. Similar effects were observed following treatment with proTAME, an inhibitor of both APC/CFzr and APC/CCdc20. Combinations of proTAME with topoisomerase inhibitors, etoposide and doxorubicin, significantly increased cell death in MM cell lines and primary cells, particularly if TOPIIα levels were first increased through pre-treatment with proTAME. Similarly, combinations of proTAME with the microtubule inhibitor vincristine resulted in enhanced cell death. This study demonstrates the potential of targeting the APC/C and its cofactors as a therapeutic approach in MM.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/genetics , Cdh1 Proteins/genetics , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Anaphase-Promoting Complex-Cyclosome/metabolism , Antineoplastic Agents/pharmacology , Cdc20 Proteins/antagonists & inhibitors , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Cdh1 Proteins/antagonists & inhibitors , Cdh1 Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Profiling , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , RNA Interference
5.
J Cell Commun Signal ; 9(1): 5-13, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25711903

ABSTRACT

The NOTCH pathway is an evolutionarily conserved signalling network, which is fundamental in regulating developmental processes in invertebrates and vertebrates (Gazave et al. in BMC Evol Biol 9:249, 2009). It regulates self-renewal (Butler et al. in Cell Stem Cell 6:251-264, 2010), differentiation (Auderset et al. in Curr Top Microbiol Immunol 360:115-134, 2012), proliferation (VanDussen et al. in Development 139:488-497, 2012) and apoptosis (Cao et al. in APMIS 120:441-450, 2012) of diverse cell types at various stages of their development. NOTCH signalling governs cell-cell interactions and the outcome of such responses is highly context specific. This makes it impossible to generalize about NOTCH functions as it stimulates survival and differentiation of certain cell types, whereas inhibiting these processes in others (Meier-Stiegen et al. in PLoS One 5:e11481, 2010). NOTCH was first identified in 1914 in Drosophila and was named after the indentations (notches) present in the wings of the mutant flies (Bigas et al. in Int J Dev Biol 54:1175-1188, 2010). Homologs of NOTCH in vertebrates were initially identified in Xenopus (Coffman et al. in Science 249:1438-1441, 1990) and in humans NOTCH was first identified in T-Acute Lymphoblastic Leukaemia (T-ALL) (Ellisen et al. in Cell 66:649-61, 1991). NOTCH signalling is integral in neurogenesis (Mead and Yutzey in Dev Dyn 241:376-389, 2012), myogenesis (Schuster-Gossler et al. in Proc Natl Acad Sci U S A 104:537-542, 2007), haematopoiesis (Bigas et al. in Int J Dev Biol 54:1175-1188, 2010), oogenesis (Xu and Gridley in Genet Res Int 2012:648207, 2012), differentiation of intestinal cells (Okamoto et al. in Am J Physiol Gastrointest Liver Physiol 296:G23-35, 2009) and pancreatic cells (Apelqvist et al. in Nature 400:877-881, 1999). The current review will focus on NOTCH signalling in normal and malignant blood cell production or haematopoiesis.

6.
J Pathol ; 231(3): 378-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24308033

ABSTRACT

Deregulated NOTCH1 has been reported in lymphoid leukaemia, although its role in chronic myeloid leukaemia (CML) is not well established. We previously reported BCR-ABL down-regulation of a novel haematopoietic regulator, CCN3, in CML; CCN3 is a non-canonical NOTCH1 ligand. This study characterizes the NOTCH1­CCN3 signalling axis in CML. In K562 cells, BCR-ABL silencing reduced full-length NOTCH1 (NOTCH1-FL) and inhibited the cleavage of NOTCH1 intracellular domain (NOTCH1-ICD), resulting in decreased expression of the NOTCH1 targets c-MYC and HES1. K562 cells stably overexpressing CCN3 (K562/CCN3) or treated with recombinant CCN3(rCCN3) showed a significant reduction in NOTCH1 signalling (> 50% reduction in NOTCH1-ICD, p < 0.05).Gamma secretase inhibitor (GSI), which blocks NOTCH1 signalling, reduced K562/CCN3 colony formation but increased that of K562/control cells. GSI combined with either rCCN3 or imatinib reduced K562 colony formation with enhanced reduction of NOTCH1 signalling observed with combination treatments. We demonstrate an oncogenic role for NOTCH1 in CML and suggest that BCR-ABL disruption of NOTCH1­CCN3 signalling contributes to the pathogenesis of CML.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Transfection
7.
Blood Rev ; 27(6): 297-304, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24183816

ABSTRACT

The ubiquitin proteasome system (UPS) plays a central role in cellular protein homeostasis through the targeted destruction of damaged/misfolded proteins and regulatory proteins that control critical cellular functions. The UPS comprises a sequential series of enzymatic activities to covalently attach ubiquitin to proteins to target them for degradation through the proteasome. Aberrancies within this system have been associated with transformation and tumourigenesis and thus, the UPS represents an attractive target for the development of anti-cancer therapies. The use of the first-in-class proteasome inhibitor, bortezomib, in the treatment of Plasma Cell Myeloma and Mantle Cell Lymphoma has validated the UPS as a therapeutic target. Following on its success, efforts are focused on the development of second-generation proteasome inhibitors and small molecule inhibitors of other components of the UPS. This review will provide an overview of the UPS and discuss current and novel therapies targeting the UPS.


Subject(s)
Hematologic Neoplasms/enzymology , Hematologic Neoplasms/therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/therapeutic use , Ubiquitin/metabolism , Hematologic Neoplasms/drug therapy , Humans , Proteasome Inhibitors/pharmacology , Ubiquitin/antagonists & inhibitors
8.
J Cell Commun Signal ; 6(1): 27-35, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21773872

ABSTRACT

CCN3, a tumour suppressor gene, is down-regulated as a result of BCR-ABL tyrosine kinase activity in Chronic Myeloid Leukaemia (CML). We have established a stable CCN3 expression model in the human K562 CML cell line and have further validated the role for CCN3 in the leukaemogenic process. K562 cells stably transfected with CCN3 (K562/CCN3; 2.25 × 10(6) copies per 50 ng cDNA) demonstrated over 50% reduction in cell growth in comparison to cells stably transfected with empty vector (K562/control; p = 0.005). K562/CCN3 cells had reduced colony formation capacity (reduced by 29.7%, p = 0.03) and reduced mitogenic signalling in comparison to K562/control cells (reduced by 29.5% (p = 0.002) and 37.4% (p = 0.017) for phosphorylation levels of ERK and AKT respectively). K562/CCN3 cells showed an accumulation of events within the subG(0) phase of the cell cycle and increased apoptosis was confirmed by a three-fold increase in annexin V binding (p < 0.05). K562/CCN3 cells exposed to Imatinib (1 µM and 5 µM) showed an increase in events within the subG(0) phase of cell cycle over 96 h and mirrored the enhanced cell kill demonstrated by Annexin staining. Wild type K562 cells treated with recombinant human Ccn3 (10 nM) in combination with Imatinib (5 µM) also displayed enhanced cell kill (p = 0.008). K562/CCN3 cells displayed increased adhesion to matrigel™ (2.92 ± 0.52 fold increase compared to K562/control) which was commensurate with increased expression of the alpha 6 and beta 4 integrins (6.53 ± 0.47 and 1.94 ± 0.07 fold increase in gene expression respectively (n = 3, p < 0.05)). CCN3 restores cellular growth regulatory properties that are absent in CML and sensitises CML cells to imatinib induced apoptosis. CCN3 may provide novel avenues for the development of alternate therapeutic strategies.

9.
J Cell Commun Signal ; 5(3): 183-91, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21638198

ABSTRACT

Chronic Myeloid Leukaemia (CML) is a myeloproliferative disorder characterized by the expression of the oncoprotein, Bcr-Abl kinase. CCN3 normally functions as a negative growth regulator, but it is downregulated in CML, the mechanism of which is not known. MicroRNAs (miRNAs) are small non-coding RNAs, which negatively regulate protein translation by binding to the complimentary sequences of the 3' UTR of messenger RNAs. Deregulated miRNA expression has emerged as a hallmark of cancer. In CML, BCR-ABL upregulates oncogenic miRNAs and downregulates tumour suppressor miRNAs favouring leukaemic transformation. We report here that the downregulation of CCN3 in CML is mediated by BCR-ABL dependent miRNAs. Using the CML cell line K562, we profiled miRNAs, which are BCR-ABL dependent by transfecting K562 cells with anti-BCR-ABL siRNA. MiRNA expression levels were quantified using the Taqman Low Density miRNA array platform. From the miRNA target prediction databases we identified miRNAs that could potentially bind to CCN3 mRNA and reduce expression. Of these, miR-130a, miR-130b, miR-148a, miR-212 and miR-425-5p were significantly reduced on BCR-ABL knockdown, with both miR-130a and miR-130b decreasing the most within 24 h of siRNA treatment. Transfection of mature sequences of miR-130a and miR-130b individually into BCR-ABL negative HL60 cells resulted in a decrease of both CCN3 mRNA and protein. The reduction in CCN3 was greatest with overexpression of miR-130a whereas miR-130b overexpression resulted only in marginal repression of CCN3. This study shows that miRNAs modulate CCN3 expression. Deregulated miRNA expression initiated by BCR-ABL may be one mechanism of downregulating CCN3 whereby leukaemic cells evade negative growth regulation.

10.
J Cell Commun Signal ; 5(2): 101-10, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21484190

ABSTRACT

The ubiquitin proteasome pathway plays a critical role in regulating many processes in the cell which are important for tumour cell growth and survival. Inhibition of proteasome function has emerged as a powerful strategy for anti-cancer therapy. Clinical validation of the proteasome as a therapeutic target was achieved with bortezomib and has prompted the development of a second generation of proteasome inhibitors with improved pharmacological properties. This review summarises the main mechanisms of action of proteasome inhibitors in cancer, the development of proteasome inhibitors as therapeutic agents and the properties and progress of next generation proteasome inhibitors in the clinic.

11.
Br J Haematol ; 153(2): 179-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21382019

ABSTRACT

Molecular testing for the BCR-ABL1 fusion gene by real time quantitative polymerase chain reaction (RT-qPCR) is the most sensitive routine approach for monitoring the response to therapy of patients with chronic myeloid leukaemia. In the context of tyrosine kinase inhibitor (TKI) therapy, the technique is most appropriate for patients who have achieved complete cytogenetic remission and can be used to define specific therapeutic milestones. To achieve this effectively, standardization of the laboratory procedures and the interpretation of results are essential. We present here consensus best practice guidelines for RT-qPCR testing, data interpretation and reporting that have been drawn up and agreed by a consortium of 21 testing laboratories in the United Kingdom and Ireland in accordance with the procedures of the UK Clinical Molecular Genetics Society.


Subject(s)
Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Monitoring, Physiologic/methods , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Ireland , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Molecular Biology , Practice Guidelines as Topic , Societies, Medical , United Kingdom
12.
J Leukoc Biol ; 88(4): 807-11, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20643815

ABSTRACT

Lymphocytes have long been established to play an important role in the regulation of hematopoiesis and produce many cytokines that act on hematopoietic progenitor cells. Previous studies by our group have shown that normal, unstimulated lymphocytes produce a protein that inhibits normal bone marrow GM colony formation. Adiponectin is an adipokine that has been demonstrated to act as a negative regulator of hematopoiesis and immune function. This study aimed to determine if the inhibitory molecule that we described previously was adiponectin. Here, we show transcription, translation, and secretion of adiponectin from lymphocytes and demonstrate that its receptors, AdipoR1 and AdipoR2, are expressed by bone marrow MNCs. We show that although the adiponectin expression is low in lymphocytes, it is sufficient to induce a significant inhibitory effect on GM precursors (CFU-GM) and activate the AMPK pathway in these cells. The regulation of adiponectin production by lymphocytes and its detailed function in suppressing GM colony formation need to be elucidated now. Our findings suggest a functional role for adiponectin as a negative regulator of granulopoiesis.


Subject(s)
Adiponectin/metabolism , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Leukopoiesis/physiology , Lymphocytes/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Granulocytes/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Immunoblotting , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Blood ; 115(11): 2241-50, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20068223

ABSTRACT

Chronic myeloid leukemia (CML) is treated effectively with tyrosine kinase inhibitors (TKIs); however, 2 key problems remain-the insensitivity of CML stem and progenitor cells to TKIs and the emergence of TKI-resistant BCR-ABL mutations. BCR-ABL activity is associated with increased proteasome activity and proteasome inhibitors (PIs) are cytotoxic against CML cell lines. We demonstrate that bortezomib is antiproliferative and induces apoptosis in chronic phase (CP) CD34+ CML cells at clinically achievable concentrations. We also show that bortezomib targets primitive CML cells, with effects on CD34+38(-), long-term culture-initiating (LTC-IC) and nonobese diabetic/severe combined immunodeficient (NOD/SCID) repopulating cells. Bortezomib is not selective for CML cells and induces apoptosis in normal CD34+38(-) cells. The effects against CML cells are seen when bortezomib is used alone and in combination with dasatinib. Bortezomib causes proteasome but not BCR-ABL inhibition and is also effective in inhibiting proteasome activity and inducing apoptosis in cell lines expressing BCR-ABL mutations, including T315I. By targeting both TKI-insensitive stem and progenitor cells and TKI-resistant BCR-ABL mutations, we believe that bortezomib offers a potential therapeutic option in CML. Because of known toxicities, including myelosuppression, the likely initial clinical application of bortezomib in CML would be in resistant and advanced disease.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Cell Culture Techniques/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Pyrazines/pharmacology , Animals , Antigens, CD34/metabolism , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Drug Synergism , Fusion Proteins, bcr-abl/metabolism , Humans , Mice , Mice, SCID , Mutant Proteins/metabolism , Proteasome Inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , Time Factors , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
14.
J Cell Commun Signal ; 3(2): 115-24, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19623482

ABSTRACT

Chronic Myeloid Leukaemia (CML) is characterized by expression of the constitutively active Bcr-Abl tyrosine kinase. We have shown previously that the negative growth regulator, CCN3, is down-regulated as a result of Bcr-Abl kinase activity and that CCN3 has a reciprocal relationship of expression with BCR-ABL. We now show that CCN3 confers growth regulation in CML cells by causing growth inhibition and regaining sensitivity to the induction of apoptosis. The mode of CCN3 induced growth regulation was investigated in K562 CML cells using gene transfection and treatment with recombinant CCN3. Both strategies showed CCN3 regulated CML cell growth by reducing colony formation capacity, increasing apoptosis and reducing ERK phosphorylation. K562 cells stably transfected to express CCN3 showed enhanced apoptosis in response to treatment with the tyrosine kinase inhibitor, imatinib. Whilst CCN3 expression was low or undetectable in CML stem cells, primary CD34+ CML progenitors were responsive to treatment with recombinant CCN3. This study shows that CCN3 is an important growth regulator in haematopoiesis, abrogation of CCN3 expression enhances BCR-ABL dependent leukaemogenesis. CCN3 restores growth regulation, regains sensitivity to the induction of apoptosis and enhances imatinib cell kill in CML cells. CCN3 may provide an additional therapeutic strategy in the management of CML.

15.
J Cell Commun Signal ; 3(2): 147-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19370401

ABSTRACT

We have adapted the CyQuant(R) assay to provide a simple, rapid, sensitive and highly reproducible method for measuring cell adhesion. The modified CyQuant(R) assay eliminates the requirement for labour intensive fluorescent labelling protocols prior to experimentation and has the sensitivity to measure small numbers (>1000) of adherent cells.

16.
Exp Hematol ; 37(3): 357-66, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19157685

ABSTRACT

OBJECTIVE: We have previously demonstrated that proteasome activity is higher in bone marrow from patients with chronic myeloid leukemia (CML) than normal controls. This study investigates whether there is any relationship between Bcr-Abl expression and proteasome activity. MATERIALS AND METHODS: Fluorogenic substrate assays and an activity-based probe were used to profile proteasome activity in CML cell-line models and the effect of the proteasome inhibitor BzLLLCOCHO on these cell-line models and primary CML cells was investigated. RESULTS: We have demonstrated that oncogenic transformation by BCR-ABL is associated with an increase in proteasome proteolytic activity. Furthermore, small interfering RNA targeted against BCR-ABL reduces proteasome activity. In addition, we have found that Bcr-Abl-positive cells are more sensitive than Bcr-Abl-negative cells to induction of apoptosis by the proteasome inhibitor BzLLLCOCHO, and that sequential addition of imatinib followed by BzLLLCOCHO has an additive effect on the induction of apoptosis in Bcr-Abl-positive cells. Finally, we demonstrate that cell lines that become resistant to imatinib remain sensitive to proteasome inhibition. CONCLUSION: This is the first time that a direct relationship has been demonstrated between BCR-ABL transformation and the enzymatic activity of the proteasome. Our results suggest that the proteasome might provide a useful therapeutic target in CML, particularly in those patients who have developed resistance to conventional treatment.


Subject(s)
Fusion Proteins, bcr-abl/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Proteasome Endopeptidase Complex/metabolism , Apoptosis , Benzamides , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Imatinib Mesylate , K562 Cells , Piperazines/pharmacology , Protease Inhibitors/pharmacology , Pyrimidines/pharmacology , Tumor Cells, Cultured
18.
Hematol Oncol ; 26(2): 73-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18324639

ABSTRACT

Many cellular processes converge on the proteasome, and its key regulatory role is increasingly being recognized. Proteasome inhibition allows the manipulation of many cellular pathways including apoptotic and cell cycle mechanisms. The proteasome inhibitor bortezomib has enhanced responses in newly diagnosed patients with myeloma and provides a new line of therapy in relapsed and refractory patients. Malignant cells are more sensitive to proteasome inhibition than normal haematopoietic cells. Proteasome inhibition enhances many conventional therapies and its role in leukaemia is promising.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia/metabolism , Leukemia/therapy , Proteasome Endopeptidase Complex/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Boronic Acids/therapeutic use , Bortezomib , Cell Cycle , Clinical Trials as Topic , Humans , Medical Oncology/methods , Models, Biological , Protease Inhibitors/therapeutic use , Proteasome Endopeptidase Complex/chemistry , Pyrazines/therapeutic use , Recurrence
20.
Cancer Res ; 66(12): 6379-86, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778216

ABSTRACT

The 26S proteasome is a multicatalytic protease responsible for regulated intracellular protein degradation. Its function is mediated by three main catalytic activities: (a) chymotrypsin-like (CT-L), (b) trypsin-like, and (c) peptidylglutamyl peptide hydrolysing (PGPH). Proteasome inhibition is an emerging therapy for many cancers and is a novel treatment for multiple myeloma. Here, we profile the contributions of the three catalytic activities in multiple myeloma cell lines and compare the specificity and cytotoxicity of the novel proteasome inhibitor BzLLLCOCHO and inhibitors PS-341 (Velcade, bortezomib) and MG-132. Using fluorogenic substrates and an active site-directed probe specific for proteasome catalytic subunits, we show differential subunit specificity for each of the inhibitors. Addition of BzLLLCOCHO strongly inhibited all three catalytic activities, treatment with PS-341 completely inhibited CT-L and PGPH activities, and treatment with MG-132 resulted in weak inhibition of the CT-L and PGPH activities. Multiple myeloma cells were more sensitive to induction of apoptosis by PS-341 and MG-132 than BzLLLCOCHO. This study emphasizes the need for further investigation of the effects of these compounds on gene and protein expression in the cell to allow for the development of more specific and targeted inhibitors.


Subject(s)
Protease Inhibitors/pharmacology , Proteasome Inhibitors , Apoptosis/drug effects , Apoptosis/physiology , Boronic Acids/pharmacology , Bortezomib , Catalysis , Cell Line, Tumor , Glyoxal/analogs & derivatives , Glyoxal/pharmacology , HeLa Cells , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Leupeptins/pharmacology , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex , Pyrazines/pharmacology , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...