Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Cell ; 137(5): 873-86, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19446321

ABSTRACT

The mTORC1 and mTORC2 pathways regulate cell growth, proliferation, and survival. We identify DEPTOR as an mTOR-interacting protein whose expression is negatively regulated by mTORC1 and mTORC2. Loss of DEPTOR activates S6K1, Akt, and SGK1, promotes cell growth and survival, and activates mTORC1 and mTORC2 kinase activities. DEPTOR overexpression suppresses S6K1 but, by relieving feedback inhibition from mTORC1 to PI3K signaling, activates Akt. Consistent with many human cancers having activated mTORC1 and mTORC2 pathways, DEPTOR expression is low in most cancers. Surprisingly, DEPTOR is highly overexpressed in a subset of multiple myelomas harboring cyclin D1/D3 or c-MAF/MAFB translocations. In these cells, high DEPTOR expression is necessary to maintain PI3K and Akt activation and a reduction in DEPTOR levels leads to apoptosis. Thus, we identify a novel mTOR-interacting protein whose deregulated overexpression in multiple myeloma cells represents a mechanism for activating PI3K/Akt signaling and promoting cell survival.


Subject(s)
Cell Survival , Multiple Myeloma/metabolism , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Line , Cyclin D1/metabolism , Cyclin D3 , Cyclins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 111(21): 7729-34, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821809

ABSTRACT

We used the I-SceI endonuclease to produce DNA double-strand breaks (DSBs) and observed that a fraction of these DSBs were repaired by insertion of sequences, which we termed "templated sequence insertions" (TSIs), derived from distant regions of the genome. These TSIs were derived from genic, retrotransposon, or telomere sequences and were not deleted from the donor site in the genome, leading to the hypothesis that they were derived from reverse-transcribed RNA. Cotransfection of RNA and an I-SceI expression vector demonstrated insertion of RNA-derived sequences at the DNA-DSB site, and TSIs were suppressed by reverse-transcriptase inhibitors. Both observations support the hypothesis that TSIs were derived from RNA templates. In addition, similar insertions were detected at sites of DNA DSBs induced by transcription activator-like effector nuclease proteins. Whole-genome sequencing of myeloma cell lines revealed additional TSIs, demonstrating that repair of DNA DSBs via insertion was not restricted to experimentally produced DNA DSBs. Analysis of publicly available databases revealed that many of these TSIs are polymorphic in the human genome. Taken together, these results indicate that insertional events should be considered as alternatives to gross chromosomal rearrangements in the interpretation of whole-genome sequence data and that this mutagenic form of DNA repair may play a role in genetic disease, exon shuffling, and mammalian evolution.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/genetics , Mutagenesis, Insertional/genetics , Retroelements/genetics , Telomere/genetics , Cell Line, Tumor , Cinnamates , Computational Biology , DNA Copy Number Variations , DNA Primers/genetics , Genetic Vectors/genetics , Humans , Hygromycin B/analogs & derivatives , Polymerase Chain Reaction
3.
Genes Chromosomes Cancer ; 55(11): 890-901, 2016 11.
Article in English | MEDLINE | ID: mdl-27311012

ABSTRACT

Multiple myeloma can be divided into two distinct genetic subgroups: hyperdiploid (HRD) or nonhyperdiploid (NHRD) myeloma. Myeloma cell lines are important tools to study myeloma cell biology and are commonly used for preclinical screening and testing of new drugs. With few exceptions human myeloma cell lines are derived from NHRD patients, even though about half of the patients have HRD myeloma. Thus, there is a need for cell lines of HRD origin to enable more representative preclinical studies. Here, we present two novel myeloma cell lines, VOLIN and KJON. Both of them were derived from patients with HRD disease and shared the same genotype as their corresponding primary tumors. The cell lines' chromosomal content, genetic aberrations, gene expression, immunophenotype as well as some of their growth characteristics are described. Neither of the cell lines was found to harbor immunoglobulin heavy chain translocations. The VOLIN cell line was established from a bone marrow aspirate and KJON from peripheral blood. We propose that these unique cell lines may be used as tools to increase our understanding of myeloma cell biology. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Line, Tumor , Multiple Myeloma/pathology , Aneuploidy , Diploidy , Humans , Immunophenotyping , Multiple Myeloma/genetics , Translocation, Genetic
4.
Cancer Cell ; 13(2): 85-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242508

ABSTRACT

Efforts to create a mouse model that provides even a phenocopy of human multiple myeloma (MM) have been unsuccessful. In this issue of Cancer Cell, Bergsagel and colleagues describe an apparent solution to this problem by creating a model in which a MYC transgene containing a stop codon and flanking Ig kappa regulatory sequences is activated sporadically in germinal center B cells by AID-dependent somatic hypermutation that reverts the stop codon. Although much remains to be done to fully characterize this model, this approach is likely to impact the creation of sporadic models for other kinds of germinal center B cell tumors.


Subject(s)
Cytidine Deaminase/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-myc/genetics , Animals , Disease Models, Animal , Disease Progression , Germinal Center/pathology , Humans , Mice , Multiple Myeloma/immunology , Paraproteinemias/pathology , Plasma Cells/enzymology , Plasma Cells/pathology
5.
Blood ; 132(6): 554-555, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30093382
6.
Cancer Cell ; 12(2): 115-30, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17692804

ABSTRACT

Mechanisms of constitutive NF-kappaB signaling in multiple myeloma are unknown. An inhibitor of IkappaB kinase beta (IKKbeta) targeting the classical NF-kappaB pathway was lethal to many myeloma cell lines. Several cell lines had elevated expression of NIK due to genomic alterations or protein stabilization, while others had inactivating mutations of TRAF3; both kinds of abnormality triggered the classical and alternative NF-kappaB pathways. A majority of primary myeloma patient samples and cell lines had elevated NF-kappaB target gene expression, often associated with genetic or epigenetic alteration of NIK, TRAF3, CYLD, BIRC2/BIRC3, CD40, NFKB1, or NFKB2. These data demonstrate that addiction to the NF-kappaB pathway is frequent in myeloma and suggest that IKKbeta inhibitors hold promise for the treatment of this disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , NF-kappa B/genetics , Signal Transduction , Baculoviral IAP Repeat-Containing 3 Protein , Blotting, Western , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cells, Cultured , Deubiquitinating Enzyme CYLD , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Plasmids , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Transfection , Translocation, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases , NF-kappaB-Inducing Kinase
7.
Genes Chromosomes Cancer ; 53(6): 467-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24585545

ABSTRACT

Primary IGH translocations involving seven recurrent partner loci and oncogenes are present in about 40% of multiple myeloma tumors. Secondary IGH rearrangements, which occur in a smaller fraction of tumors, usually are complex structures, including insertions or translocations that can involve three chromosomes, and often with involvement of MYC. The main approach to detect IGH rearrangements is interphase-but sometimes metaphase-FISH strategies that use a telomeric variable region probe and a centromeric constant region/ Eα enhancer or 3' flanking probe to detect a separation of these two probes, or a fusion of these probes with probes located at nonrandom partner sites in the genome. We analyzed 18 myeloma cell lines for detection discrepancies among Vysis, Cytocell, and in-house IGH probe sets that hybridize with differing sequences in the IGH locus. There were no detection discrepancies for the three telomeric IGH probes, or for unrearranged IGH loci or primary IGH translocations using the centromeric IGH probes. However, the majority of complex IGH rearrangements had detection discrepancies among the three centromeric IGH probes.


Subject(s)
DNA Probes/chemistry , Gene Rearrangement , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Cell Line, Tumor , Chromosomes, Human/genetics , Humans , In Situ Hybridization, Fluorescence , Interphase , Multiple Myeloma/pathology , Mutagenesis, Insertional , Translocation, Genetic
8.
Blood ; 120(2): 238-40, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22791771

ABSTRACT

In this issue of Blood, Chesi et al show that their genetically engineered mouse model of multiple myeloma can predict positive or negative activity of drugs previously tested in clinical trials.

9.
Blood ; 120(12): 2351-2, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22996653

ABSTRACT

In this issue of Blood, Holien et al report that MYC addiction is responsible for rapid death of myeloma cell lines and primary myeloma tumor cells treated with a specific MYC inhibitor.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-myc/metabolism , Humans
10.
J Exp Med ; 204(3): 633-43, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17353367

ABSTRACT

To elucidate the mechanisms underlying chromosomal translocations in diffuse large B cell lymphoma (DLBCL), we investigated the nature and extent of immunoglobulin class switch recombination (CSR) in these tumors. We used Southern blotting to detect legitimate and illegitimate CSR events in tumor samples of the activated B cell-like (ABC), germinal center B cell-like (GCB), and primary mediastinal B cell lymphoma (PMBL) subgroups of DLBCL. The frequency of legitimate CSR was lower in ABC DLBCL than in GCB DLBCL and PMBL. In contrast, ABC DLBCL had a higher frequency of internal deletions within the switch mu (Smu) region compared with GCB DLBCL and PMBL. ABC DLBCLs also had frequent deletions within Sgamma and other illegitimate switch recombinations. Sequence analysis revealed ongoing Smu deletions within ABC DLBCL tumor clones, which were accompanied by ongoing duplications and activation-induced cytidine deaminase-dependent somatic mutations. Unexpectedly, short fragments derived from multiple chromosomes were interspersed within Smu in one case. These findings suggest that ABC DLBCLs have abnormalities in the regulation of CSR that could predispose to chromosomal translocations. Accordingly, aberrant switch recombination was responsible for translocations in ABC DLBCLs involving BCL6, MYC, and a novel translocation partner, SPIB.


Subject(s)
Immunoglobulin Class Switching/immunology , Lymphocyte Activation/genetics , Lymphoma, B-Cell/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Recombination, Genetic , Translocation, Genetic , Cell Line, Tumor , Humans , Immunoglobulin Class Switching/genetics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/immunology , Lymphoma, Large B-Cell, Diffuse/genetics , Tumor Cells, Cultured
11.
Blood ; 117(8): 2396-404, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21163924

ABSTRACT

Modulating aberrant transcription of oncogenes is a relatively unexplored opportunity in cancer therapeutics. In approximately 10% of multiple myelomas, the initiating oncogenic event is translocation of musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a transcriptional activator of key target genes, including cyclinD2. Our prior work showed that MAF is up-regulated in an additional 30% of multiple myeloma cases. The present study describes a common mechanism inducing MAF transcription in both instances. The second mode of MAF transcription occurred in myelomas with multiple myeloma SET domain (MMSET) translocation. MMSET knockdown decreased MAF transcription and cell viability. A small-molecule screen found an inhibitor of mitogen-activated protein kinase kinase (MEK), which activates extracellular signal-regulated kinase (ERK)-MAP kinases, reduced MAF mRNA in cells representing MMSET or MAF subgroups. ERK activates transcription of FOS, part of the AP-1 transcription factor. By chromatin immunoprecipitation, FOS bound the MAF promoter, and MEK inhibition decreased this interaction. MEK inhibition selectively induced apoptosis in MAF-expressing myelomas, and FOS inactivation was similarly toxic. Reexpression of MAF rescued cells from death induced by MMSET depletion, MEK inhibition, or FOS inactivation. The data presented herein demonstrate that the MEK-ERK pathway regulates MAF transcription, providing molecular rationale for clinical evaluation of MEK inhibitors in MAF-expressing myeloma.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Multiple Myeloma/drug therapy , Oncogene Protein v-maf/genetics , Protein Kinases/therapeutic use , Apoptosis , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/physiology , Humans , Repressor Proteins/physiology , Transcription, Genetic
12.
Nat Rev Cancer ; 2(3): 175-87, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11990854

ABSTRACT

Multiple myeloma is a neoplasm of terminally differentiated B cells (plasma cells) in which chromosome translocations frequently place oncogenes under the control of immunoglobulin enhancers. Unlike most haematopoietic cancers, multiple myeloma often has complex chromosomal abnormalities that are reminiscent of epithelial tumours. What causes full-blown myeloma? And can our molecular understanding of this common haematological malignancy be used to develop effective preventive and treatment strategies?


Subject(s)
Multiple Myeloma/genetics , B-Lymphocytes/physiology , Enhancer Elements, Genetic , Humans , Immunoglobulins/genetics , Multiple Myeloma/pathology , Neoplasm Staging , Oncogenes , Translocation, Genetic
13.
Blood ; 115(17): 3541-52, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20053756

ABSTRACT

Mutations involving the nuclear factor-kappaB (NF-kappaB) pathway are present in at least 17% of multiple myeloma (MM) tumors and 40% of MM cell lines (MMCLs). These mutations, which are apparent progression events, enable MM tumors to become less dependent on bone marrow signals that activate NF-kappaB. Studies on a panel of 51 MMCLs provide some clarification of the mechanisms through which these mutations act and the significance of classical versus alternative activation of NF-kappaB. First, only one mutation (NFKB2) selectively activates the alternative pathway, whereas several mutations (CYLD, NFKB1, and TACI) selectively activate the classical pathway. However, most mutations affecting NF-kappaB-inducing kinase (NIK) levels (NIK, TRAF2, TRAF3, cIAP1&2, and CD40) activate the alternative but often both pathways. Second, we confirm the critical role of TRAF2 in regulating NIK degradation, whereas TRAF3 enhances but is not essential for cIAP1/2-mediated proteasomal degradation of NIK in MM. Third, using transfection to selectively activate the classical or alternative NF-kappaB pathways, we show virtually identical changes in gene expression in one MMCL, whereas the changes are similar albeit nonidentical in a second MMCL. Our results suggest that MM tumors can achieve increased autonomy from the bone marrow microenvironment by mutations that activate either NF-kappaB pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Deubiquitinating Enzyme CYLD , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Multiple Myeloma , Mutation , NF-kappa B p50 Subunit/genetics , NF-kappa B p52 Subunit/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases , NF-kappaB-Inducing Kinase
14.
Cancer Cell ; 5(2): 191-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14998494

ABSTRACT

The oncogene c-maf is translocated in approximately 5%-10% of multiple myelomas. Unexpectedly, we observed c-maf expression in myeloma cell lines lacking c-maf translocations and in 50% of multiple myeloma bone marrow samples. By gene expression profiling, we identified three c-maf target genes: cyclin D2, integrin beta7, and CCR1. c-maf transactivated the cyclin D2 promoter and enhanced myeloma proliferation, whereas dominant inhibition of c-maf blocked tumor formation in immunodeficient mice. c-maf-driven expression of integrin beta7 enhanced myeloma adhesion to bone marrow stroma and increased production of VEGF. We propose that c-maf transforms plasma cells by stimulating cell cycle progression and by altering bone marrow stromal interactions. The frequent overexpression of c-maf in myeloma makes it an attractive target for therapeutic intervention.


Subject(s)
Bone Marrow/metabolism , DNA-Binding Proteins/metabolism , Multiple Myeloma/metabolism , Plasma Cells/metabolism , Proto-Oncogene Proteins/metabolism , Stromal Cells/metabolism , Animals , Bone Marrow/physiopathology , Cadherins/metabolism , Cell Adhesion/physiology , Cyclin D2 , Cyclins/metabolism , Gene Expression Profiling , Humans , Integrin beta Chains/metabolism , Mice , Models, Animal , Multiple Myeloma/physiopathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Plasma Cells/cytology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-maf , Stromal Cells/cytology , Transplantation, Heterologous/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
15.
Blood ; 113(22): 5418-22, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19234139

ABSTRACT

Preexisting plasma cell disorders, monoclonal gammopathy of undetermined significance, or smoldering myeloma are present in at least one-third of multiple myeloma patients. However, the proportion of patients with a preexisting plasma cell disorder has never been determined by laboratory testing on prediagnostic sera. We cross-referenced our autologous stem cell transplantation database with the Department of Defense Serum Repository. Serum protein electrophoresis, immunofixation electrophoresis, and serum free light-chain analysis were performed on all sera collected 2 or more years before diagnosis to detect a monoclonal gammopathy (M-Ig). In 30 of 90 patients, 110 prediagnostic samples were available from 2.2 to 15.3 years before diagnosis. An M-Ig was detected initially in 27 of 30 patients (90%, 95% confidence interval, 74%-97%); by serum protein electrophoresis and/or immunofixation electrophoresis in 21 patients (77.8%), and only by serum free light-chain analysis in 6 patients (22.2%). Four patients had only one positive sample within 4 years before diagnosis, with all preceding sera negative. All 4 patients with light-chain/nonsecretory myeloma evolved from a light-chain M-Ig. A preexisting M-Ig is present in most multiple myeloma patients before diagnosis. Some patients progress rapidly through a premalignant phase. Light-chain detected M-Ig is a new entity that requires further study.


Subject(s)
Multiple Myeloma/etiology , Paraproteinemias/complications , Precancerous Conditions/complications , Precancerous Conditions/diagnosis , Adult , Aged , Cohort Studies , Early Detection of Cancer , Female , Humans , Immunoglobulins/analysis , Immunoglobulins/blood , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/diagnosis , Paraproteinemias/blood , Paraproteinemias/diagnosis , Precancerous Conditions/blood , Time Factors
16.
DNA Repair (Amst) ; 8(3): 330-5, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19064000

ABSTRACT

The Karpas-620 human myeloma cell line (HMCL) expresses high levels of Cyclin D1 (CCND1), but has a der(8)t(8;11) and a der(14)t(8;14), and not a conventional t(11;14). Fluorescent in situ hybridization (FISH) and array comparative genomic hybridization (aCGH) studies suggest that der(14)t(11;14) from a primary translocation underwent a secondary translocation with chromosome 8 to generate der(8)t(8;[14];11) and der(14)t(8;[11];14). Both secondary derivatives share extensive identical sequences from chromosomes 8, 11, and 14, including MYC and the 3' IgH enhancers. Der(14), with MYC located approximately 700 kb telomeric to the 3'IGH enhancer, expresses MYC. By contrast, der(8), with both CCND1 and MYC repositioned near a 3'IGH enhancer, expresses CCND1, which is telomeric of the enhancer, but not MYC, which is centromeric to the enhancer. The secondary translocation that dysregulated MYC resulted in extensive regions from both donor chromosomes being transmitted to both derivative chromosomes, suggesting a defect in DNA recombination or repair in the myeloma tumor cell.


Subject(s)
Cell Line, Tumor , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 8/genetics , Cyclin D1/metabolism , Enhancer Elements, Genetic , Multiple Myeloma , Proto-Oncogene Proteins c-myc/metabolism , Translocation, Genetic , Animals , Comparative Genomic Hybridization , Cyclin D1/genetics , Gene Dosage/genetics , Humans , In Situ Hybridization, Fluorescence , Mice , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA/metabolism
17.
Blood Cancer Discov ; 1(3): 221-223, 2020 11.
Article in English | MEDLINE | ID: mdl-34661146

ABSTRACT

A comprehensive genomic analysis of structural variants in multiple myeloma in this issue highlights the key role of these events, involving primarily the immunoglobulin heavy chain locus in disease initiation and the MYC locus in disease progression. However, the current study reveals the large number of genomic hotspots, oncogenes, tumor suppressor genes, and recombination mechanisms that contribute to multiple myeloma heterogeneity. See related article by Rustad et al., p. 258.

18.
Genes Chromosomes Cancer ; 47(7): 573-90, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18381641

ABSTRACT

The pathogenesis of multiple myeloma (MM) is thought to involve at least two pathways, which generate hyperdiploid (HRD) or nonhyperdiploid (NHRD) tumors, respectively. Apart from chromosome content, the two pathways are distinguished by five primary immunoglobulin heavy chain (IGH) rearrangements (4p16, FGFR3, and MMSET; 6p21, CCND3; 11q13, CCND1; 16q23, MAF; 20q12, MAFB) that are present mainly in NHRD tumors. To determine the prevalence and structures of IGH, immunoglobulin (IG) light chain, and MYC genomic rearrangements in MM, we have done comprehensive metaphase fluorescent in situ hybridization analyses on 48 advanced MM tumors and 47 MM cell lines. As expected, the prevalence of the five primary IGH rearrangements was nearly 70% in NHRD tumors, but only 12% in HRD tumors. However, IGH rearrangements not involving one of the five primary partners, and IG light chain rearrangements, have a similar prevalence in HRD and NHRD tumors. In addition, MYC rearrangements, which are thought to be late progression events that sometimes do not involve an IG heavy or light chain locus, also have a similar prevalence in HRD and NHRD tumors. In contrast to the primary IGH rearrangements, which usually are simple balanced translocations, these other IG rearrangements usually have complex structures, as previously described for MYC rearrangements in MM. We conclude that IG light chain and MYC rearrangements, as well as secondary IGH rearrangements, make similar contributions to the progression of both HRD and NHRD MM tumors.


Subject(s)
Gene Rearrangement , Genes, Immunoglobulin Heavy Chain/genetics , Genes, myc/genetics , Immunoglobulin Light Chains/genetics , Multiple Myeloma/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Diploidy , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Multiple Myeloma/pathology , Translocation, Genetic , Tumor Cells, Cultured
19.
J Natl Cancer Inst Monogr ; (39): 25-31, 2008.
Article in English | MEDLINE | ID: mdl-18647998

ABSTRACT

Translocations involving an MYC gene (c >> N >>L) are very late tumor progression events and provide a paradigm for secondary translocations in multiple myeloma. Using a combination of fluorescent in situ hybridization and comparative genomic hybridization arrays (aCGH), we have identified rearrangements of an MYC gene in 40 of 43 independent myeloma cell lines. A majority of MYC translocations involve an Ig locus (IgH > Iglambda >> Igkappa), but the breakpoints only infrequently occur near or within switch regions or V(D)J sequences. Surprisingly, about 40% of MYC translocations do not involve an Ig locus. The MYC translocations mostly are nonreciprocal translocations or insertions, often with the involvement of three chromosomes and sometimes with associated duplication, amplification, inversion, and other associated chromosomal abnormalities. High-density aCGH analyses should facilitate the cloning of MYC breakpoints, enabling the determination of their structures and perhaps elucidating how rearrangements not involving an Ig gene cause dysregulation of an MYC gene.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 8/genetics , Genes, myc/genetics , Multiple Myeloma/genetics , Translocation, Genetic , Gene Expression Profiling , Gene Rearrangement/genetics , Genes, Immunoglobulin Heavy Chain , Humans , Immunoglobulin Light Chains , In Situ Hybridization, Fluorescence , Karyotyping , Nucleic Acid Hybridization , Tumor Cells, Cultured
20.
Mol Cancer Ther ; 16(9): 2008-2021, 2017 09.
Article in English | MEDLINE | ID: mdl-28522584

ABSTRACT

Cancer treatments often require combinations of molecularly targeted agents to be effective. mTORi (rapamycin) and HDACi (MS-275/entinostat) inhibitors have been shown to be effective in limiting tumor growth, and here we define part of the cooperative action of this drug combination. More than 60 human cancer cell lines responded synergistically (CI<1) when treated with this drug combination compared with single agents. In addition, a breast cancer patient-derived xenograft, and a BCL-XL plasmacytoma mouse model both showed enhanced responses to the combination compared with single agents. Mice bearing plasma cell tumors lived an average of 70 days longer on combination treatment compared with single agents. A set of 37 genes cooperatively affected (34 downregulated; 3 upregulated) by the combination responded pharmacodynamically in human myeloma cell lines, xenografts, and a P493 model, and were both enriched in tumors, and correlated with prognostic markers in myeloma patient datasets. Genes downregulated by the combination were overexpressed in several untreated cancers (breast, lung, colon, sarcoma, head and neck, myeloma) compared with normal tissues. The MYC/E2F axis, identified by upstream regulator analyses and validated by immunoblots, was significantly inhibited by the drug combination in several myeloma cell lines. Furthermore, 88% of the 34 genes downregulated have MYC-binding sites in their promoters, and the drug combination cooperatively reduced MYC half-life by 55% and increased degradation. Cells with MYC mutations were refractory to the combination. Thus, integrative approaches to understand drug synergy identified a clinically actionable strategy to inhibit MYC/E2F activity and tumor cell growth in vivoMol Cancer Ther; 16(9); 2008-21. ©2017 AACR.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Repair , DNA Replication/drug effects , Disease Models, Animal , Drug Synergism , Female , Gene Expression Profiling , Humans , Mice , Pharmacogenetics , Pharmacogenomic Variants , Protein Stability , Proteolysis , Transcriptome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL