Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Med ; 29(3): 632-645, 2023 03.
Article in English | MEDLINE | ID: mdl-36928817

ABSTRACT

The historical lack of preclinical models reflecting the genetic heterogeneity of multiple myeloma (MM) hampers the advance of therapeutic discoveries. To circumvent this limitation, we screened mice engineered to carry eight MM lesions (NF-κB, KRAS, MYC, TP53, BCL2, cyclin D1, MMSET/NSD2 and c-MAF) combinatorially activated in B lymphocytes following T cell-driven immunization. Fifteen genetically diverse models developed bone marrow (BM) tumors fulfilling MM pathogenesis. Integrative analyses of ∼500 mice and ∼1,000 patients revealed a common MAPK-MYC genetic pathway that accelerated time to progression from precursor states across genetically heterogeneous MM. MYC-dependent time to progression conditioned immune evasion mechanisms that remodeled the BM microenvironment differently. Rapid MYC-driven progressors exhibited a high number of activated/exhausted CD8+ T cells with reduced immunosuppressive regulatory T (Treg) cells, while late MYC acquisition in slow progressors was associated with lower CD8+ T cell infiltration and more abundant Treg cells. Single-cell transcriptomics and functional assays defined a high ratio of CD8+ T cells versus Treg cells as a predictor of response to immune checkpoint blockade (ICB). In clinical series, high CD8+ T/Treg cell ratios underlie early progression in untreated smoldering MM, and correlated with early relapse in newly diagnosed patients with MM under Len/Dex therapy. In ICB-refractory MM models, increasing CD8+ T cell cytotoxicity or depleting Treg cells reversed immunotherapy resistance and yielded prolonged MM control. Our experimental models enable the correlation of MM genetic and immunological traits with preclinical therapy responses, which may inform the next-generation immunotherapy trials.


Subject(s)
Multiple Myeloma , Mice , Animals , Multiple Myeloma/therapy , Multiple Myeloma/drug therapy , CD8-Positive T-Lymphocytes , Immune Evasion , T-Lymphocytes, Regulatory , Immunotherapy/adverse effects , Tumor Microenvironment/genetics
2.
Blood ; 141(9): 1047-1059, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36455198

ABSTRACT

Venetoclax combination therapies are becoming the standard of care in acute myeloid leukemia (AML). However, the therapeutic benefit of these drugs in older/unfit patients is limited to only a few months, highlighting the need for more effective therapies. Protein phosphatase 2A (PP2A) is a tumor suppressor phosphatase with pleiotropic functions that becomes inactivated in ∼70% of AML cases. PP2A promotes cancer cell death by modulating the phosphorylation state in a variety of proteins along the mitochondrial apoptotic pathway. We therefore hypothesized that pharmacological PP2A reactivation could increase BCL2 dependency in AML cells and, thus, potentiate venetoclax-induced cell death. Here, by using 3 structurally distinct PP2A-activating drugs, we show that PP2A reactivation synergistically enhances venetoclax activity in AML cell lines, primary cells, and xenograft models. Through the use of gene editing tools and pharmacological approaches, we demonstrate that the observed therapeutic synergy relies on PP2A complexes containing the B56α regulatory subunit, of which expression dictates response to the combination therapy. Mechanistically, PP2A reactivation enhances venetoclax-driven apoptosis through simultaneous inhibition of antiapoptotic BCL2 and extracellular signal-regulated kinase signaling, with the latter decreasing MCL1 protein stability. Finally, PP2A targeting increases the efficacy of the clinically approved venetoclax and azacitidine combination in vitro, in primary cells, and in an AML patient-derived xenograft model. These preclinical results provide a scientific rationale for testing PP2A-activating drugs with venetoclax combinations in AML.


Subject(s)
Leukemia, Myeloid, Acute , Protein Phosphatase 2 , Humans , Aged , Myeloid Cell Leukemia Sequence 1 Protein , Cell Line, Tumor , Proto-Oncogene Proteins c-bcl-2 , Leukemia, Myeloid, Acute/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Apoptosis
3.
Cancer Discov ; 11(5): 1268-1285, 2021 05.
Article in English | MEDLINE | ID: mdl-33355179

ABSTRACT

For millions of years, endogenous retroelements have remained transcriptionally silent within mammalian genomes by epigenetic mechanisms. Modern anticancer therapies targeting the epigenetic machinery awaken retroelement expression, inducing antiviral responses that eliminate tumors through mechanisms not completely understood. Here, we find that massive binding of epigenetically activated retroelements by RIG-I and MDA5 viral sensors promotes ATP hydrolysis and depletes intracellular energy, driving tumor killing independently of immune signaling. Energy depletion boosts compensatory ATP production by switching glycolysis to mitochondrial oxidative phosphorylation, thereby reversing the Warburg effect. However, hyperfunctional succinate dehydrogenase in mitochondrial electron transport chain generates excessive oxidative stress that unleashes RIP1-mediated necroptosis. To maintain ATP generation, hyperactive mitochondrial membrane blocks intrinsic apoptosis by increasing BCL2 dependency. Accordingly, drugs targeting BCL2 family proteins and epigenetic inhibitors yield synergistic responses in multiple cancer types. Thus, epigenetic therapy kills cancer cells by rewiring mitochondrial metabolism upon retroelement activation, which primes mitochondria to apoptosis by BH3-mimetics. SIGNIFICANCE: The state of viral mimicry induced by epigenetic therapies in cancer cells remodels mitochondrial metabolism and drives caspase-independent tumor cell death, which sensitizes to BCL2 inhibitor drugs. This novel mechanism underlies clinical efficacy of hypomethylating agents and venetoclax in acute myeloid leukemia, suggesting similar combination therapies for other incurable cancers.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Mitochondria/drug effects , Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Humans
4.
Blood ; 137(1): 49-60, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32693406

ABSTRACT

Patients with multiple myeloma (MM) carrying standard- or high-risk cytogenetic abnormalities (CAs) achieve similar complete response (CR) rates, but the later have inferior progression-free survival (PFS). This questions the legitimacy of CR as a treatment endpoint and represents a biological conundrum regarding the nature of tumor reservoirs that persist after therapy in high-risk MM. We used next-generation flow (NGF) cytometry to evaluate measurable residual disease (MRD) in MM patients with standard- vs high-risk CAs (n = 300 and 90, respectively) enrolled in the PETHEMA/GEM2012MENOS65 trial, and to identify mechanisms that determine MRD resistance in both patient subgroups (n = 40). The 36-month PFS rates were higher than 90% in patients with standard- or high-risk CAs achieving undetectable MRD. Persistent MRD resulted in a median PFS of âˆ¼3 and 2 years in patients with standard- and high-risk CAs, respectively. Further use of NGF to isolate MRD, followed by whole-exome sequencing of paired diagnostic and MRD tumor cells, revealed greater clonal selection in patients with standard-risk CAs, higher genomic instability with acquisition of new mutations in high-risk MM, and no unifying genetic event driving MRD resistance. Conversely, RNA sequencing of diagnostic and MRD tumor cells uncovered the selection of MRD clones with singular transcriptional programs and reactive oxygen species-mediated MRD resistance in high-risk MM. Our study supports undetectable MRD as a treatment endpoint for patients with MM who have high-risk CAs and proposes characterizing MRD clones to understand and overcome MRD resistance. This trial is registered at www.clinicaltrials.gov as #NCT01916252.


Subject(s)
Drug Resistance, Neoplasm/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm, Residual/pathology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boron Compounds/therapeutic use , Bortezomib/therapeutic use , Chromosome Aberrations , Dexamethasone/therapeutic use , Female , Flow Cytometry , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Lenalidomide/therapeutic use , Male , Middle Aged , Progression-Free Survival , Treatment Outcome
5.
J Pathol ; 245(1): 61-73, 2018 05.
Article in English | MEDLINE | ID: mdl-29464716

ABSTRACT

The increased risk of Richter transformation (RT) in patients with chronic lymphocytic leukaemia (CLL) due to Epstein-Barr virus (EBV) reactivation during immunosuppressive therapy with fludarabine other targeted agents remains controversial. Among 31 RT cases classified as diffuse large B-cell lymphoma (DLBCL), seven (23%) showed EBV expression. In contrast to EBV- tumours, EBV+ DLBCLs derived predominantly from IGVH-hypermutated CLL, and they also showed CLL-unrelated IGVH sequences more frequently. Intriguingly, despite having different cellular origins, clonally related and unrelated EBV+ DLBCLs shared a previous history of immunosuppressive chemo-immunotherapy, a non-germinal centre DLBCL phenotype, EBV latency programme type II or III, and very short survival. These data suggested that EBV reactivation during therapy-related immunosuppression can transform either CLL cells or non-tumoural B lymphocytes into EBV+ DLBCL. To investigate this hypothesis, xenogeneic transplantation of blood cells from 31 patients with CLL and monoclonal B-cell lymphocytosis (MBL) was performed in Rag2-/- IL2γc-/- mice. Remarkably, the recipients' impaired immunosurveillance favoured the spontaneous outgrowth of EBV+ B-cell clones from 95% of CLL and 64% of MBL patients samples, but not from healthy donors. Eventually, these cells generated monoclonal tumours (mostly CLL-unrelated but also CLL-related), recapitulating the principal features of EBV+ DLBCL in patients. Accordingly, clonally related and unrelated EBV+ DLBCL xenografts showed indistinguishable cellular, virological and molecular features, and synergistically responded to combined inhibition of EBV replication with ganciclovir and B-cell receptor signalling with ibrutinib in vivo. Our study underscores the risk of RT driven by EBV in CLL patients receiving immunosuppressive therapies, and provides the scientific rationale for testing ganciclovir and ibrutinib in EBV+ DLBCL. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Herpesvirus 4, Human/drug effects , Immunosuppressive Agents/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Adult , Aged , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/pathology , Female , Herpesvirus 4, Human/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Male , Middle Aged
6.
Blood ; 123(26): 4111-9, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24786774

ABSTRACT

Acquired resistance to targeted drugs is emerging as an obstacle to successful cancer treatment. Recently, a BCL2-selective BH3 mimetic termed ABT-199 showed promising therapeutic results in BCL2-dependent tumors. Based on its high affinity for BCL2, we studied potential mechanisms conferring resistance upon ABT-199 therapy, aiming to anticipate its occurrence in the clinic. Two models of resistant lymphomas were established by continuous ABT-199 exposure. In resistant Bcl2-expressing mouse lymphoma cells, 2 missense mutations within the Bcl2 BH3 domain were identified. Both F101C and F101L mutations impeded ABT-199 binding to the BH3 domain, therefore suppressing mitochondrial apoptosis. In resistant human lymphoma cells, a missense mutation in the C-terminal transmembrane domain of proapoptotic BAX (G179E) was found, which abrogated BAX anchoring to mitochondria and blocked ABT-199-induced apoptosis both in vitro and in vivo. Importantly, G179E BAX mutation also induced partial cross-resistance to other antineoplastic drugs. Our study reveals the acquisition of mutations in BCL2 family proteins as a novel mechanism of apoptosis resistance in cancer. These results anticipate the potential development of such mutations in patients treated with ABT-199, providing a basis to preventing their occurrence and to designing drugs able to circumvent the acquired resistance.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/drug effects , Lymphoma/metabolism , Mutation, Missense , Sulfonamides/pharmacology , bcl-2-Associated X Protein/metabolism , Amino Acid Substitution , Animals , Antineoplastic Agents , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , bcl-2-Associated X Protein/genetics
7.
Blood ; 121(21): 4311-20, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23580662

ABSTRACT

B-cell maturation and germinal center (GC) formation are dependent on the interplay between BCL6 and other transcriptional regulators. FOXP1 is a transcription factor that regulates early B-cell development, but whether it plays a role in mature B cells is unknown. Analysis of human tonsillar B-cell subpopulations revealed that FOXP1 shows the opposite expression pattern to BCL6, suggesting that FOXP1 regulates the transition from resting follicular B cell to activated GC B cell. Chromatin immunoprecipitation-on-chip and gene expression assays on B cells indicated that FOXP1 acts as a transcriptional activator and repressor of genes involved in the GC reaction, half of which are also BCL6 targets. To study FOXP1 function in vivo, we developed transgenic mice expressing human FOXP1 in lymphoid cells. These mice exhibited irregular formation of splenic GCs, showing a modest increase in naïve and marginal-zone B cells and a significant decrease in GC B cells. Furthermore, aberrant expression of FOXP1 impaired transcription of noncoding γ1 germline transcripts and inhibited efficient class switching to the immunoglobulin G1 isotype. These studies show that FOXP1 is physiologically downregulated in GC B cells and that aberrant expression of FOXP1 impairs mechanisms triggered by B-cell activation, potentially contributing to B-cell lymphomagenesis.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , Germinal Center/cytology , Lymphoma/immunology , Repressor Proteins/metabolism , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Down-Regulation/immunology , Forkhead Transcription Factors/immunology , Germinal Center/immunology , Humans , Lymphoma/metabolism , Mice , Mice, Transgenic , Palatine Tonsil/cytology , Proto-Oncogene Proteins c-bcl-6 , Repressor Proteins/immunology , Transcriptional Activation/immunology
8.
Br J Haematol ; 158(6): 712-26, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22816737

ABSTRACT

Using high-resolution genomic microarray analysis, a distinct genomic profile was defined in 114 samples from patients with splenic marginal zone lymphoma (SMZL). Deletion or uniparental disomy of chromosome 7q were detected in 42 of 114 (37%) SMZLs but in only nine of 170 (5%) mature B-cell lymphomas (P < 0·00001). The presence of unmutated IGHV, genomic complexity, 17p13-TP53 deletion and 8q-MYC gain, but not 7q deletion, correlated with shorter overall survival of SMZL patients. Mapping studies narrowed down a commonly deleted region of 2·7 Mb in 7q32.1-q32.2 spanning a region between the SND1 and COPG2 genes. High-throughput sequencing analysis of the 7q32-deleted segment did not identify biallelic deletions/insertions or clear pathogenic gene mutations, but detected six nucleotide changes in IRF5 (n = 2), TMEM209 (n = 2), CALU (n = 1) and ZC3HC1 (n = 1) not found in healthy individuals. Comparative expression analysis found a fourfold down-regulation of IRF5 gene in lymphomas with 7q32 deletion versus non-deleted tumours (P = 0·032). Ectopic expression of IRF5 in marginal-zone lymphoma cells decreased proliferation and increased apoptosis in vitro, and impaired lymphoma development in vivo. These results show that cryptic deletions, insertions and/or point mutations inactivating genes within 7q32 are not common in SMZL, and suggest that IRF5 may be a haploinsufficient tumour suppressor in this lymphoma entity.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Genes, Tumor Suppressor , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Interferon Regulatory Factors/genetics , Lymphoma, B-Cell, Marginal Zone/genetics , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Sequence Deletion , Splenic Neoplasms/genetics , Animals , Apoptosis/genetics , Cell Division/drug effects , Cell Line, Tumor/transplantation , Chromosomes, Human, Pair 7/ultrastructure , Comparative Genomic Hybridization , Gene Expression Regulation, Neoplastic , Genes, Immunoglobulin , Humans , Interferon Regulatory Factors/biosynthesis , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/physiology , Kaplan-Meier Estimate , Lymphoma, B-Cell, Marginal Zone/mortality , Lymphoma, B-Cell, Marginal Zone/pathology , Mice , Mice, Knockout , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Point Mutation , Real-Time Polymerase Chain Reaction , Splenic Neoplasms/mortality , Splenic Neoplasms/pathology , Translocation, Genetic
9.
Proc Natl Acad Sci U S A ; 108(30): 12461-6, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21746927

ABSTRACT

The chromosomal translocation t(11;14)(q13;q32) leading to cyclin-D1 overexpression plays an essential role in the development of mantle cell lymphoma (MCL), an aggressive tumor that remains incurable with current treatment strategies. Cyclin-D1 has been postulated as an effective therapeutic target, but the evaluation of this target has been hampered by our incomplete understanding of its oncogenic functions and by the lack of valid MCL murine models. To address these issues, we generated a cyclin-D1-driven mouse model in which cyclin-D1 expression can be regulated externally. These mice developed cyclin-D1-expressing lymphomas capable of recapitulating features of human MCL. We found that cyclin-D1 inactivation was not sufficient to induce lymphoma regression in vivo; however, using a combination of in vitro and in vivo assays, we identified a novel prosurvival cyclin-D1 function in MCL cells. Specifically, we found that cyclin-D1, besides increasing cell proliferation through deregulation of the cell cycle at the G(1)-S transition, sequestrates the proapoptotic protein BAX in the cytoplasm, thereby favoring BCL2's antiapoptotic function. Accordingly, cyclin-D1 inhibition sensitized the lymphoma cells to apoptosis through BAX release. Thus, genetic or pharmacologic targeting of cyclin-D1 combined with a proapoptotic BH3 mimetic synergistically killed the cyclin-D1-expressing murine lymphomas, human MCL cell lines, and primary lymphoma cells. Our study identifies a role of cyclin-D1 in deregulating apoptosis in MCL cells, and highlights the potential benefit of simultaneously targeting cyclin-D1 and survival pathways in patients with MCL. This effective combination therapy also might be exploited in other cyclin-D1-expressing tumors.


Subject(s)
Cyclin D1/metabolism , Lymphoma, Mantle-Cell/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Survival , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Disease Models, Animal , Gene Amplification , Genes, bcl-2 , Humans , Lymphoma, Mantle-Cell/etiology , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/therapy , Mice , Nitrophenols/pharmacology , Piperazines/pharmacology , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/antagonists & inhibitors
10.
Haematologica ; 96(7): 980-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21459790

ABSTRACT

BACKGROUND: LMO2 is highly expressed at the most immature stages of lymphopoiesis. In T-lymphocytes, aberrant LMO2 expression beyond those stages leads to T-cell acute lymphoblastic leukemia, while in B cells LMO2 is also expressed in germinal center lymphocytes and diffuse large B-cell lymphomas, where it predicts better clinical outcome. The implication of LMO2 in B-cell acute lymphoblastic leukemia must still be explored. DESIGN AND METHODS: We measured LMO2 expression by real time RT-PCR in 247 acute lymphoblastic leukemia patient samples with cytogenetic data (144 of them also with survival and immunophenotypical data) and in normal hematopoietic and lymphoid cells. RESULTS: B-cell acute lymphoblastic leukemia cases expressed variable levels of LMO2 depending on immunophenotypical and cytogenetic features. Thus, the most immature subtype, pro-B cells, displayed three-fold higher LMO2 expression than pre-B cells, common-CD10+ or mature subtypes. Additionally, cases with TEL-AML1 or MLL rearrangements exhibited two-fold higher LMO2 expression compared to cases with BCR-ABL rearrangements or hyperdyploid karyotype. Clinically, high LMO2 expression correlated with better overall survival in adult patients (5-year survival rate 64.8% (42.5%-87.1%) vs. 25.8% (10.9%-40.7%), P= 0.001) and constituted a favorable independent prognostic factor in B-ALL with normal karyotype: 5-year survival rate 80.3% (66.4%-94.2%) vs. 63.0% (46.1%-79.9%) (P= 0.043). CONCLUSIONS: Our data indicate that LMO2 expression depends on the molecular features and the differentiation stage of B-cell acute lymphoblastic leukemia cells. Furthermore, assessment of LMO2 expression in adult patients with a normal karyotype, a group which lacks molecular prognostic factors, could be of clinical relevance.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Metalloproteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Child , Child, Preschool , Humans , Immunophenotyping , Infant , Karyotyping , LIM Domain Proteins , Middle Aged , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prognosis , Proto-Oncogene Proteins , Survival Analysis , Treatment Outcome , Young Adult
11.
Blood ; 116(14): 2531-42, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20570860

ABSTRACT

In Burkitt lymphoma/leukemia (BL), achievement of complete remission with first-line chemotherapy remains a challenging issue, as most patients who respond remain disease-free, whereas those refractory have few options of being rescued with salvage therapies. The mechanisms underlying BL chemoresistance and how it can be circumvented remain undetermined. We previously reported the frequent inactivation of the proapoptotic BIM gene in B-cell lymphomas. Here we show that BIM epigenetic silencing by concurrent promoter hypermethylation and deacetylation occurs frequently in primary BL samples and BL-derived cell lines. Remarkably, patients with BL with hypermethylated BIM presented lower complete remission rate (24% vs 79%; P = .002) and shorter overall survival (P = .007) than those with BIM-expressing lymphomas, indicating that BIM transcriptional repression may mediate tumor chemoresistance. Accordingly, by combining in vitro and in vivo studies of human BL-xenografts grown in immunodeficient RAG2(-/-)γc(-/-) mice and of murine B220(+)IgM(+) B-cell lymphomas generated in Eµ-MYC and Eµ-MYC-BIM(+/-) transgenes, we demonstrate that lymphoma chemoresistance is dictated by BIM gene dosage and is reversible on BIM reactivation by genetic manipulation or after treatment with histone-deacetylase inhibitors. We suggest that the combination of histone-deacetylase inhibitors and high-dose chemotherapy may overcome chemoresistance, achieve durable remission, and improve survival of patients with BL.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Drug Resistance, Neoplasm/drug effects , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Bcl-2-Like Protein 11 , Cell Line, Tumor , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured
12.
Methods Mol Biol ; 576: 231-77, 2010.
Article in English | MEDLINE | ID: mdl-19882266

ABSTRACT

During the last decade, gene expression microarrays and array-based comparative genomic hybridization (array-CGH) have unraveled the complexity of human tumor genomes more precisely and comprehensively than ever before. More recently, the simultaneous assessment of global changes in messenger RNA (mRNA) expression and in DNA copy number through "integrative oncogenomic" analyses has allowed researchers the access to results uncovered through the analysis of one-dimensional data sets, thus accelerating cancer gene discovery. In this chapter, we discuss the major contributions of DNA microarrays to the study of hematological malignancies, focusing on the integrative oncogenomic approaches that correlate genomic and transcriptomic data. We also present the basic aspects of these methodologies and their present and future application in clinical oncology.


Subject(s)
Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis/methods , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Genomics , Humans , Leukemia/genetics , Lymphoma/genetics , Multiple Myeloma/genetics , Neoplasms/genetics , Neoplasms/metabolism , Nucleic Acid Hybridization , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism
13.
Eur J Cancer ; 45(10): 1877-89, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19403302

ABSTRACT

BACKGROUND: Expression of the pro-apoptotic BCL-2-interacting mediator (BIM) has recently been implicated in imatinib-induced apoptosis of BCR-ABL1(+) cells. However, the mechanisms involved in the regulation of BIM in CML and its role in the clinical setting have not been established. DESIGN AND METHODS: We analysed the mRNA expression of BIM in 100 newly diagnosed patients with CML in chronic phase by Q-RT-PCR and the protein levels by Western blot analysis. Methylation status was analysed by bisulphite genomic sequencing and MSP. CML cell lines were treated with imatinib and 5-aza-2'-deoxycytidine, and were transfected with two different siRNAs against BIM and cell proliferation and apoptosis were analysed. RESULTS: We demonstrated that down-regulation of BIM expression was present in 36% of the patients and was significantly associated with a lack of optimal response to imatinib as indicated by the decrease in cytogenetic and molecular responses at 6, 12 and 18 months in comparison with patients with normal BIM expression (p<0.05). Expression of BIM was mediated by promoter hypermethylation as demonstrated by restoration of BIM expression after treatment of CML cells with 5-aza-2'-deoxycytidine. Using CML cell lines with low and normal expression of BIM we further demonstrated that the expression of BIM is required for imatinib-induced CML apoptosis. CONCLUSION: Our data indicate that down-regulation of BIM is epigenetically controlled by methylation in a percentage of CML patients and has an unfavourable prognostic impact, and that the combination of imatinib with a de-methylating agent may result in improved responses in patients with decreased expression of BIM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Down-Regulation/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Membrane Proteins/biosynthesis , Piperazines/pharmacology , Proto-Oncogene Proteins/biosynthesis , Pyrimidines/pharmacology , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Bcl-2-Like Protein 11 , Benzamides , Cell Proliferation/drug effects , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Evaluation, Preclinical/methods , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Membrane Proteins/genetics , Middle Aged , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tumor Cells, Cultured
14.
Biochemistry ; 46(49): 13983-93, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18001139

ABSTRACT

Cobyric acid synthetase (CbiP) from Salmonella typhimurium catalyzes the glutamine and ATP-dependent amidation of carboxylates b, d, e, and g within adenosyl cobyrinic acid a,c-diamide. After each round of catalysis the partially amidated intermediates are released into solution and the four carboxylates are amidated in the sequential order of e, d, b, and g for the wild type enzyme. In the presence of [gamma-18O4]-ATP and adenosyl cobyrinic a,c-diamide the enzyme will catalyze the positional isotope exchange of the betagamma-bridge oxygen with the two beta-nonbridge oxygens. These results support the proposal that ATP is used to activate the carboxylate groups via the formation of a phosphorylated intermediate. CbiP catalyzes the hydrolysis of glutamine in the absence of ATP or adenosyl cobyrinic acid a,c-diamide, but the rate of glutamine hydrolysis is enhanced by a factor of 60 in the presence of these two substrates together. This result suggests that the formation of the phosphorylated intermediate is coupled to the activation of the site utilized for the hydrolysis of glutamine. However, the rate of glutamine hydrolysis is approximately 2.5 times the rate of ADP formation, indicating that the two active sites are partially uncoupled from one another and that some of the ammonia from glutamine hydrolysis leaks into the bulk solution. The mutation of D146 to either alanine or asparagine results in a protein that is able to catalyze the formation of cobyric acid. However, the strict amidation order observed with the wild type CbiP is partially randomized with carboxylate b being amidated last. With the D146N mutant, the predominant pathway occurs in the sequence d, e, g, and b. It is proposed that this residue enforces the amidation order in the wild type enzyme via charge-charge repulsion between the side chain carboxylate and the carboxylates of the substrate.


Subject(s)
Transaminases/genetics , Transaminases/metabolism , Cobamides/metabolism , Kinetics , Models, Molecular , Point Mutation , Salmonella typhimurium/enzymology
16.
Blood ; 109(1): 271-80, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16960149

ABSTRACT

Integrative genomic and gene-expression analyses have identified amplified oncogenes in B-cell non-Hodgkin lymphoma (B-NHL), but the capability of such technologies to localize tumor suppressor genes within homozygous deletions remains unexplored. Array-based comparative genomic hybridization (CGH) and gene-expression microarray analysis of 48 cell lines derived from patients with different B-NHLs delineated 20 homozygous deletions at 7 chromosome areas, all of which contained tumor suppressor gene targets. Further investigation revealed that only a fraction of primary biopsies presented inactivation of these genes by point mutation or intragenic deletion, but instead some of them were frequently silenced by epigenetic mechanisms. Notably, the pattern of genetic and epigenetic inactivation differed among B-NHL subtypes. Thus, the P53-inducible PIG7/LITAF was silenced by homozygous deletion in primary mediastinal B-cell lymphoma and by promoter hypermethylation in germinal center lymphoma, the proapoptotic BIM gene presented homozygous deletion in mantle cell lymphoma and promoter hypermethylation in Burkitt lymphoma, the proapoptotic BH3-only NOXA was mutated and preferentially silenced in diffuse large B-cell lymphoma, and INK4c/P18 was silenced by biallelic mutation in mantle-cell lymphoma. Our microarray strategy has identified novel candidate tumor suppressor genes inactivated by genetic and epigenetic mechanisms that substantially vary among the B-NHL subtypes.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Lymphoma, B-Cell/genetics , Membrane Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/genetics , Sequence Deletion , Transcription Factors/genetics , Vesicular Transport Proteins/genetics , Adaptor Proteins, Signal Transducing , Apoptosis/genetics , Bcl-2-Like Protein 11 , Biopsy , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Human/genetics , Chromosomes, Human/ultrastructure , DNA Methylation , DNA Mutational Analysis , DNA, Neoplasm/genetics , Epigenesis, Genetic , Gene Dosage , Gene Expression Regulation, Neoplastic , Gene Silencing , Homozygote , Humans , Lymphoma, B-Cell/classification , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Point Mutation , Promoter Regions, Genetic/genetics , RNA-Binding Proteins , Sorting Nexins
17.
Biochemistry ; 44(19): 7115-24, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15882050

ABSTRACT

Isoaspartyl dipeptidase (IAD) is a member of the amidohydrolase superfamily and catalyzes the hydrolytic cleavage of beta-aspartyl dipeptides. Structural studies of the wild-type enzyme have demonstrated that the active site consists of a binuclear metal center positioned at the C-terminal end of a (beta/alpha)(8)-barrel domain. Steady-state kinetic parameters for the hydrolysis of beta-aspartyl dipeptides were obtained at pH 8.1. The pH-rate profiles for the hydrolysis of beta-Asp-Leu were obtained for the Zn/Zn-, Co/Co-, Ni/Ni-, and Cd/Cd-substituted forms of IAD. Bell-shaped profiles were observed for k(cat) and k(cat)/K(m) as a function of pH for all four metal-substituted forms. The pK(a) of the group that must be unprotonated for catalytic activity varied according to the specific metal ion bound in the active site, whereas the pK(a) of the group that must be protonated for catalytic activity was relatively independent of the specific metal ion present. The identity of the group that must be unprotonated for catalytic activity was consistent with the hydroxide that bridges the two divalent cations of the binuclear metal center. The identity of the group that must be protonated for activity was consistent with the free alpha-amino group of the dipeptide substrate. Kinetic constants were obtained for the mutant enzymes at conserved residues Glu77, Tyr137, Arg169, Arg233, Asp285, and Ser289. The catalytic properties of the wild-type and mutant enzymes, coupled with the X-ray crystal structure of the D285N mutant complexed with beta-Asp-His, are consistent with a chemical reaction mechanism for the hydrolysis of dipeptides that is initiated by the polarization of the amide bond via complexation to the beta-metal ion of the binuclear metal center. Nucleophilic attack by the bridging hydroxide is facilitated by abstraction of its proton by the side chain carboxylate of Asp285. Collapse of the tetrahedral intermediate and cleavage of the carbon-nitrogen bond occur with donation of a proton from the protonated form of Asp285.


Subject(s)
Dipeptidases/chemistry , Escherichia coli Proteins/chemistry , Asparagine/genetics , Aspartic Acid/genetics , Binding Sites/genetics , Catalysis , Crystallography, X-Ray , Dipeptidases/antagonists & inhibitors , Dipeptidases/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Hydrogen-Ion Concentration , Isoaspartic Acid/chemistry , Kinetics , Models, Chemical , Models, Molecular , Mutagenesis, Site-Directed , Protons , Substrate Specificity/genetics
18.
J Mol Biol ; 349(1): 127-41, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15876373

ABSTRACT

Carbamoyl phosphate synthetase I (CPSI) deficiency, a recessively inherited error of the urea cycle, causes life-threatening hyperammonaemia. CPSI is a multidomain 1500-residue liver mitochondrial matrix protein that is allosterically activated by N-acetyl-l-glutamate, and which synthesises carbamoyl phosphate (CP) in three steps: bicarbonate phosphorylation by ATP, carbamate synthesis from carboxyphosphate and ammonia, and carbamate phosphorylation by ATP. Several missense mutations of CPSI have been reported in patients with CPSI deficiency, but the actual pathogenic potential and effects on the enzyme of these mutations remain non-characterised. Since the structure of Escherichia coli CPS is known and systems for its overexpression and purification are available, we have constructed and purified eight site-directed mutants of E.coli CPS affecting the enzyme large subunit (A126M, R169H, Q262P, N301K, P360L, V640R, R675L, S789P) that are homologous to corresponding missense mutations found in patients with CPSI deficiency, studying their stability and their ability to catalyse the CPS reaction as well as the partial reactions that reflect the different reactional steps, and analysing the substrate kinetics for the overall and partial reactions. The results show that all the mutations significantly decrease CP synthesis without completely inactivating the enzyme (as reflected in the catalysis of at least one partial reaction), that one of these mutations (Q262P) causes marked enzyme instability, and validate the use of E.coli CPS as a pathogenicity testing model for CPSI deficiency. The causality of the reported clinical mutations is supported and the derangements caused by the mutations are identified, revealing the specific roles of the residues that are mutated. In particular, the findings highlight the importance for carbamate phosphorylation and for allosteric activation of a loop that coordinates K(+), stress the key role of intersubunit interactions for CPS stability, and suggest that lid opening at both phosphorylation sites is concerted.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/metabolism , Escherichia coli/enzymology , Adenosine Triphosphate/metabolism , Allosteric Regulation/physiology , Amino Acid Substitution , Bicarbonates/metabolism , Carbamates/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Escherichia coli/genetics , Humans , Mutation , Phosphorylation
19.
Biochemistry ; 43(33): 10619-27, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15311923

ABSTRACT

Cobyrinic acid a,c-diamide synthetase from Salmonella typhimurium (CbiA) is the first glutamine amidotransferase in the anaerobic biosynthetic pathway of vitamin B(12) and catalyzes the ATP-dependent synthesis of cobyrinic acid a,c-diamide from cobyrinic acid using either glutamine or ammonia as the nitrogen source. The cbiA gene was cloned, the overexpressed protein was purified to homogeneity, and the kinetic parameters were determined. CbiA is a monomer with K(m) values of 0.74, 2.7, 53, and 26 200 microM for cobyrinic acid, ATP, glutamine, and ammonia, respectively. Analysis of the glutaminase partial reaction demonstrated that the hydrolysis of glutamine and the synthesis of the cobyrinic acid a,c-diamide product are uncoupled. The time course for the synthesis of the diamide product and positional isotope exchange experiments demonstrate that CbiA catalyzes the sequential amidation of the c- and a-carboxylate groups of cobyrinic acid via the formation of a phosphorylated intermediate. These results support a model for the catalytic mechanism in which CbiA catalyzes the amidation of the c-carboxylate, and then the intermediate is released into solution and binds to the same catalytic site for the amidation of the a-carboxylate. Several conserved residues in the synthetase active site were mutated to address the molecular basis of the amidation order; however, no changes in the order of amidation were obtained. The mutants D45N, D48N, and E90Q have a dramatic effect on the catalytic activity, whereas no effect was found for the mutant D97N. The substitutions by alanine of L47 and Y46 residues specifically decrease the affinity of the enzyme for the c-monoamide intermediate.


Subject(s)
Salmonella typhimurium/enzymology , Transaminases/chemistry , Transaminases/metabolism , Amino Acid Substitution , Catalysis , Cloning, Molecular , Kinetics , Oxygen Isotopes , Phosphorylation , Substrate Specificity , Transaminases/genetics
20.
Bioorg Chem ; 32(2): 63-75, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14990305

ABSTRACT

Asparagine synthetase B (AsnB) catalyzes the formation of asparagine in an ATP-dependent reaction using glutamine or ammonia as a nitrogen source. To obtain a better understanding of the catalytic mechanism of this enzyme, we report the cloning, expression, and kinetic analysis of the glutamine- and ammonia-dependent activities of AsnB from Vibrio cholerae. Initial velocity, product inhibition, and dead-end inhibition studies were utilized in the construction of a model for the kinetic mechanism of the ammonia- and glutamine-dependent activities. The reaction sequence begins with the ordered addition of ATP and aspartate. Pyrophosphate is released, followed by the addition of ammonia and the release of asparagine and AMP. Glutamine is simultaneously hydrolyzed at a second site and the ammonia intermediate diffuses through an interdomain protein tunnel from the site of production to the site of utilization. The data were also consistent with the dead-end binding of asparagine to the glutamine binding site and PP(i) with free enzyme. The rate of hydrolysis of glutamine is largely independent of the activation of aspartate and thus the reaction rates at the two active sites are essentially uncoupled from one another.


Subject(s)
Aspartate-Ammonia Ligase/metabolism , Vibrio cholerae/enzymology , Aspartate-Ammonia Ligase/antagonists & inhibitors , Glutamine/metabolism , Hydrolysis , Kinetics , Molecular Structure , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...