Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Genome Biol ; 25(1): 143, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822412

ABSTRACT

BACKGROUND: Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence. RESULTS: Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers. CONCLUSIONS: Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Histone-Lysine N-Methyltransferase/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Myeloid-Lymphoid Leukemia Protein/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Core Binding Factor Alpha 2 Subunit/genetics
2.
Oncoimmunology ; 11(1): 2109861, 2022.
Article in English | MEDLINE | ID: mdl-35979386

ABSTRACT

Tyrosine kinase inhibitors (TKIs) have dramatically improved the survival in chronic myeloid leukemia (CML), but residual disease typically persists even after prolonged treatment. Several lines of evidence suggest that TKIs administered to CML patients upregulate interferon γ (IFNγ) production, which may counteract the anti-tumorigenic effects of the therapy. We now show that activated T cell-conditioned medium (TCM) enhanced proliferation and counteracted imatinib-induced apoptosis of CML cells, and addition of a neutralizing anti-IFNγ antibody at least partially inhibited the anti-apoptotic effect. Likewise, recombinant IFNγ also reduced imatinib-induced apoptosis of CML cells. This anti-apoptotic effect of IFNγ was independent of alternative IFNγ signaling pathways, but could be notably diminished by STAT1-knockdown. Furthermore, IFNγ upregulated the expression of several anti-apoptotic proteins, including MCL1, PARP9, and PARP14, both in untreated and imatinib-treated primary human CD34+ CML stem/progenitor cells. Our results suggest that activated T cells in imatinib-treated CML patients can directly rescue CML cells from imatinib-induced apoptosis at least partially through the secretion of IFNγ, which exerts a rapid, STAT1-dependent anti-apoptotic effect potentially through the simultaneous upregulation of several key hematopoietic survival factors. These mechanisms may have a major clinical impact, when targeting residual leukemic stem/progenitor cells in CML.


Subject(s)
Interferon-gamma , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Antigens, CD34/metabolism , Antigens, CD34/pharmacology , Apoptosis , Cell Line, Tumor , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Stem Cells/metabolism , Up-Regulation
4.
Cell Death Dis ; 12(10): 875, 2021 09 25.
Article in English | MEDLINE | ID: mdl-34564697

ABSTRACT

Tyrosine kinase inhibitor (TKI) treatment has dramatically improved the survival of chronic myeloid leukemia (CML) patients, but measurable residual disease typically persists. To more effectively eradicate leukemia cells, simultaneous targeting of BCR-ABL1 and additional CML-related survival proteins has been proposed. Notably, several highly specific myeloid cell leukemia 1 (MCL1) inhibitors have recently entered clinical trials for various hematologic malignancies, although not for CML, reflecting the insensitivity of CML cell lines to single MCL1 inhibition. Here, we show that combining TKI (imatinib, nilotinib, dasatinib, or asciminib) treatment with the small-molecule MCL1 inhibitor S63845 exerted strong synergistic antiviability and proapoptotic effects on CML lines and CD34+ stem/progenitor cells isolated from untreated CML patients in chronic phase. Using wild-type BCR-ABL1-harboring CML lines and their T315I-mutated sublines (generated by CRISPR/Cas9-mediated homologous recombination), we prove that the synergistic proapoptotic effect of the drug combination depended on TKI-mediated BCR-ABL1 inhibition, but not on TKI-related off-target mechanisms. Moreover, we demonstrate that colony formation of CML but not normal hematopoietic stem/progenitor cells became markedly reduced upon combination treatment compared to imatinib monotherapy. Our results suggest that dual targeting of MCL1 and BCR-ABL1 activity may efficiently eradicate residual CML cells without affecting normal hematopoietic stem/progenitors.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiophenes/pharmacology , Antigens, CD34/metabolism , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Clone Cells , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis/drug effects , Small Molecule Libraries/pharmacology , bcl-X Protein/metabolism
5.
Genome Med ; 12(1): 99, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33218352

ABSTRACT

BACKGROUND: Tight regulatory loops orchestrate commitment to B cell fate within bone marrow. Genetic lesions in this gene regulatory network underlie the emergence of the most common childhood cancer, acute lymphoblastic leukemia (ALL). The initial genetic hits, including the common translocation that fuses ETV6 and RUNX1 genes, lead to arrested cell differentiation. Here, we aimed to characterize transcription factor activities along the B-lineage differentiation trajectory as a reference to characterize the aberrant cell states present in leukemic bone marrow, and to identify those transcription factors that maintain cancer-specific cell states for more precise therapeutic intervention. METHODS: We compared normal B-lineage differentiation and in vivo leukemic cell states using single cell RNA-sequencing (scRNA-seq) and several complementary genomics profiles. Based on statistical tools for scRNA-seq, we benchmarked a workflow to resolve transcription factor activities and gene expression distribution changes in healthy bone marrow lymphoid cell states. We compared these to ALL bone marrow at diagnosis and in vivo during chemotherapy, focusing on leukemias carrying the ETV6-RUNX1 fusion. RESULTS: We show that lymphoid cell transcription factor activities uncovered from bone marrow scRNA-seq have high correspondence with independent ATAC- and ChIP-seq data. Using this comprehensive reference for regulatory factors coordinating B-lineage differentiation, our analysis of ETV6-RUNX1-positive ALL cases revealed elevated activity of multiple ETS-transcription factors in leukemic cells states, including the leukemia genome-wide association study hit ELK3. The accompanying gene expression changes associated with natural killer cell inactivation and depletion in the leukemic immune microenvironment. Moreover, our results suggest that the abundance of G1 cell cycle state at diagnosis and lack of differentiation-associated regulatory network changes during induction chemotherapy represent features of chemoresistance. To target the leukemic regulatory program and thereby overcome treatment resistance, we show that inhibition of ETS-transcription factors reduced cell viability and resolved pathways contributing to this using scRNA-seq. CONCLUSIONS: Our data provide a detailed picture of the transcription factor activities characterizing both normal B-lineage differentiation and those acquired in leukemic bone marrow and provide a rational basis for new treatment strategies targeting the immune microenvironment and the active regulatory network in leukemia.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia/genetics , Lymphocytes/physiology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Bone Marrow , Cell Line, Tumor , Child , Core Binding Factor Alpha 2 Subunit/metabolism , Drug Delivery Systems , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Leukemia/drug therapy , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Transcription Factors , Transcriptome , Translocation, Genetic , ETS Translocation Variant 6 Protein
6.
EMBO J ; 35(20): 2192-2212, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27625374

ABSTRACT

SOX9 is a master transcription factor that regulates development and stem cell programs. However, its potential oncogenic activity and regulatory mechanisms that control SOX9 protein stability are poorly understood. Here, we show that SOX9 is a substrate of FBW7, a tumor suppressor, and a SCF (SKP1/CUL1/F-box)-type ubiquitin ligase. FBW7 recognizes a conserved degron surrounding threonine 236 (T236) in SOX9 that is phosphorylated by GSK3 kinase and consequently degraded by SCFFBW7α Failure to degrade SOX9 promotes migration, metastasis, and treatment resistance in medulloblastoma, one of the most common childhood brain tumors. FBW7 is either mutated or downregulated in medulloblastoma, and in cases where FBW7 mRNA levels are low, SOX9 protein is significantly elevated and this phenotype is associated with metastasis at diagnosis and poor patient outcome. Transcriptional profiling of medulloblastoma cells expressing a degradation-resistant SOX9 mutant reveals activation of pro-metastatic genes and genes linked to cisplatin resistance. Finally, we show that pharmacological inhibition of PI3K/AKT/mTOR pathway activity destabilizes SOX9 in a GSK3/FBW7-dependent manner, rendering medulloblastoma cells sensitive to cytostatic treatment.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Medulloblastoma/metabolism , SOX9 Transcription Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Aniline Compounds/pharmacology , Animals , Benzamides , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement , Chromones/pharmacology , Cisplatin/pharmacology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Humans , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Mice, Nude , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , SOX9 Transcription Factor/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
7.
EMBO Mol Med ; 5(7): 1067-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23776131

ABSTRACT

SCF (Skp1/Cul1/F-box) ubiquitin ligases act as master regulators of cellular homeostasis by targeting key proteins for ubiquitylation. Here, we identified a hitherto uncharacterized F-box protein, FBXO28 that controls MYC-dependent transcription by non-proteolytic ubiquitylation. SCF(FBXO28) activity and stability are regulated during the cell cycle by CDK1/2-mediated phosphorylation of FBXO28, which is required for its efficient ubiquitylation of MYC and downsteam enhancement of the MYC pathway. Depletion of FBXO28 or overexpression of an F-box mutant unable to support MYC ubiquitylation results in an impairment of MYC-driven transcription, transformation and tumourigenesis. Finally, in human breast cancer, high FBXO28 expression and phosphorylation are strong and independent predictors of poor outcome. In conclusion, our data suggest that SCF(FBXO28) plays an important role in transmitting CDK activity to MYC function during the cell cycle, emphasizing the CDK-FBXO28-MYC axis as a potential molecular drug target in MYC-driven cancers, including breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast/pathology , CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Amino Acid Sequence , Breast/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Phosphorylation , Prognosis , Promoter Regions, Genetic , Proteolysis , SKP Cullin F-Box Protein Ligases/analysis , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , Survival Analysis , Transcriptional Activation , Ubiquitination
8.
Cell Cycle ; 12(6): 889-98, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23422002

ABSTRACT

Neuroblastoma is the most common solid tumor in childhood and represents 15% of all children's cancer deaths. We have previously demonstrated that tripartite motif 16 (TRIM16), a member of the RING B-box coiled-coil (RBCC)/tripartite totif (TRIM) protein family, has significant effects on neuroblastoma proliferation and migration in vitro and tumorigenicity in vivo. However, the mechanism by which this putative tumor suppressor influences cell proliferation and tumorigenicity was undetermined. Here we show, for the first time, TRIM16's striking pattern of expression and dynamic localization during cell cycle progression and neuroblastoma tumor development. In a tyrosine hydroxylase MYCN (TH-MYCN) neuroblastoma mouse model, immunohistochemical staining revealed strong nuclear TRIM16 expression in differentiating ganglia cells but not in the tumor-initiating cells. Furthermore in vitro studies clearly demonstrated that during G 1 cell cycle phase, TRIM16 protein expression is upregulated and shifts to the nucleus of cells. TRIM16 also plays a role in cell cycle progression through changes in Cyclin D1 and p27 expression. Importantly, using TRIM16 deletion mutants, an uncharacterized protein domain of TRIM16 was found to be required for both TRIM16's growth inhibitory effects and its nuclear localization. Taken together, our data suggest that TRIM16 acts as a novel regulator of both neuroblastoma G 1/S progression and cell differentiation.


Subject(s)
Cell Nucleus/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA-Binding Proteins/metabolism , Neuroblastoma/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/genetics , G1 Phase , G1 Phase Cell Cycle Checkpoints , HEK293 Cells , Humans , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , RNA Interference , RNA, Small Interfering , Transcription Factors/genetics , Tripartite Motif Proteins , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin-Protein Ligases
9.
PLoS One ; 7(5): e37470, 2012.
Article in English | MEDLINE | ID: mdl-22629402

ABSTRACT

The TRIM family of proteins is distinguished by its tripartite motif (TRIM). Typically, TRIM proteins contain a RING finger domain, one or two B-box domains, a coiled-coil domain and the more variable C-terminal domains. TRIM16 does not have a RING domain but does harbour two B-box domains. Here we showed that TRIM16 homodimerized through its coiled-coil domain and heterodimerized with other TRIM family members; TRIM24, Promyelocytic leukaemia (PML) protein and Midline-1 (MID1). Although, TRIM16 has no classic RING domain, three-dimensional modelling of TRIM16 suggested that its B-box domains adopts RING-like folds leading to the hypothesis that TRIM16 acts as an ubiquitin ligase. Consistent with this hypothesis, we demonstrated that TRIM16, devoid of a classical RING domain had auto-polyubiquitination activity and acted as an E3 ubiquitin ligase in vivo and in vitro assays. Thus via its unique structure, TRIM16 possesses both heterodimerization function with other TRIM proteins and also has E3 ubiquitin ligase activity.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Models, Molecular , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Structure, Tertiary/genetics , Transcription Factors/genetics , Transfection , Tripartite Motif Proteins , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
10.
J Pathol ; 226(3): 451-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009481

ABSTRACT

Retinoid therapy is used for chemo-prevention in immuno-suppressed patients at high risk of developing skin cancer. The retinoid signalling molecule, tripartite motif protein 16 (TRIM16), is a regulator of keratinocyte differentiation and a tumour suppressor in retinoid-sensitive neuroblastoma. We sought to determine the role of TRIM16 in skin squamous cell carcinoma (SCC) pathogenesis. We have shown that TRIM16 expression was markedly reduced during the histological progression from normal skin to actinic keratosis and SCC. SCC cell lines exhibited lower cytoplasmic and nuclear TRIM16 expression compared with primary human keratinocyte (PHK) cells due to reduced TRIM16 protein stability. Overexpressed TRIM16 translocated to the nucleus, inducing growth arrest and cell differentiation. In SCC cells, TRIM16 bound to and down regulated nuclear E2F1, this is required for cell replication. Retinoid treatment increased nuclear TRIM16 expression in retinoid-sensitive PHK cells, but not in retinoid-resistant SCC cells. Overexpression of TRIM16 reduced SCC cell migration, which required the C-terminal RET finger protein (RFP)-like domain of TRIM16. The mesenchymal intermediate filament protein, vimentin, was directly bound and down-regulated by TRIM16 and was required for TRIM16-reduced cell migration. Taken together, our data suggest that loss of TRIM16 expression plays an important role in the development of cutaneous SCC and is a determinant of retinoid sensitivity.


Subject(s)
Carcinoma, Squamous Cell/etiology , DNA-Binding Proteins/metabolism , Skin Neoplasms/etiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Movement/physiology , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Dermatologic Agents/pharmacology , Down-Regulation , Humans , Immunohistochemistry , In Vitro Techniques , Isotretinoin/pharmacology , Protein Binding , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Tripartite Motif Proteins , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Vimentin/metabolism
11.
Cell Cycle ; 7(8): 1075-82, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18414042

ABSTRACT

The ubiquitin-mediated turnover of cyclin E is regulated by phosphorylation and the activity of the ubiquitin ligase SCF(Cdc4) (also known as SCF(Fbw7)). In 293A cells, SCF complexes containing two different Cdc4 isoforms, alpha and gamma, are required for efficient cyclin E ubiquitylation. Whereas SCF(Cdc4gamma) ubiquitylates cyclin E directly, SCF(Cdc4alpha) serves as a cofactor for Pin1-mediated prolyl isomerization of the cyclin E phosphodegron, essential to potentiate ubiquitylation. In the current study, we show that the requirement for both Cdc4alpha and gamma is general, except in cell lines where cyclin E is expressed at an elevated level. Under these circumstances, Cdc4alpha is sufficient for cyclin E turnover. Furthermore, the requirement for Cdc4gamma can be bypassed by ectopic overexpression of cyclin E.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin E/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , DNA Primers/genetics , F-Box-WD Repeat-Containing Protein 7 , Fluorescent Antibody Technique , Humans , Phosphorylation , Protein Isoforms/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
12.
Cancer Res ; 67(12): 5611-6, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575125

ABSTRACT

Notch signaling is of crucial importance in normal T-cell development and Notch 1 is frequently mutated in T-cell acute lymphoblastic leukemias (T-ALL), leading to aberrantly high Notch signaling. In this report, we determine whether T-ALL mutations occur not only in Notch1 but also in the F-box protein hCdc4 (Sel-10, Ago, or Fbxw7), a negative regulator of Notch1. We show that the hCDC4 gene is mutated in leukemic cells from more than 30% of patients with pediatric T-ALL and derived cell lines. Most hCDC4 mutations found were missense substitutions at critical arginine residues (Arg(465), Arg(479), and Arg(505)) localized in the substrate-binding region of hCdc4. Cells inactivated for hCdc4 and T-ALL cells containing hCDC4 mutations exhibited an increased Notch1 protein half-life, consistent with the proposed role of hCdc4 in ubiquitin-dependent proteolysis of Notch1. Furthermore, restoration of wild-type but not mutant hCdc4 in HCT 116 hCDC4-negative cells led to an increased Notch1 ubiquitylation and decreased Notch1 signaling. These results show that hCdc4 mutations interfere with normal Notch1 regulation in vivo. Finally, we found that mutations in hCDC4 and NOTCH1 can occur in the same cancers and that patients carrying hCDC4 and/or NOTCH1 mutations have a favorable overall survival. Collectively, these data show that mutation of hCDC4 is a frequent event in T-ALL and suggest that hCDC4 mutations and gain-of-function mutations in NOTCH1 might synergize in contributing to the development of pediatric T-ALL leukemogenesis.


Subject(s)
Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Receptor, Notch1/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Base Sequence , Blotting, Western , Cell Cycle Proteins/metabolism , Child , DNA Mutational Analysis , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Female , Humans , Male , Mutation , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Oncogene ; 23(35): 5941-9, 2004 Aug 05.
Article in English | MEDLINE | ID: mdl-15208682

ABSTRACT

RASSF1A, a major member of the RASSF1 gene family, is silenced by promoter methylation at a high frequency in a large number of human solid tumors. Controlled expression of RASSF1A reverts the tumorigenic phenotype of several human cancer cell lines. Here we investigated another main isoform, RASSF1C, and compared it with RASSF1A in the gene inactivation test (GIT), based on a tetracycline regulation system. In the small-cell lung cancer (SCLC) line U2020, only RASSF1A has shown growth inhibitory activity in vitro, while in the prostate cell line LNCaP and renal cell carcinoma (RCC) line KRC/Y both RASSF1A and RASSF1C showed similar (approximately 90%) suppressing activity in vitro. Both RASSF1C and RASSF1A suppressed the tumorigenicity of the KRC/Y RCC cell line in SCID mice. Mutations, deletions and loss of expression of RASSF1A and RASSF1C transgenes were identified in all 15 grown SCID tumors. In contrast, the mutant RASSF1A containing Cys65Arg and Val211Ala had reduced growth suppression activity both in vitro and in vivo and did not show any further changes in four grown SCID tumors. In addition, RASSF1C was shown to induce cell cycle arrest in KRC/Y cells. These results strongly imply that like RASSF1A the RASSF1C gene could serve a tumor suppressor function.


Subject(s)
Genes, Tumor Suppressor , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle , Cell Line, Tumor , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Male , Mice , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , Tumor Suppressor Proteins/physiology
14.
Proc Natl Acad Sci U S A ; 101(14): 4906-11, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15051889

ABSTRACT

Chromosome 3p21.3 region is frequently (>90%) deleted in lung and other major human carcinomas. We subdivided 3p21.3 into LUCA and AP20 subregions and discovered frequent homozygous deletions (10-18%) in both subregions. This finding strongly implies that they harbor multiple tumor suppressor genes involved in the origin and/or development of major epithelial cancers. In this study, we performed an initial analysis of RBSP3/HYA22, a candidate tumor suppressor genes located in the AP20 region. Two sequence splice variants of RBSP3/HYA22 (A and B) were identified, and we provide evidence for their tumor suppressor function. By sequence analysis RBSP3/HYA22 belongs to a gene family of small C-terminal domain phosphatases that may control the RNA polymerase II transcription machinery. Expression of the gene was drastically (>20-fold) decreased in 11 of 12 analyzed carcinoma cell lines and in three of eight tumor biopsies. We report missense and nonsense mutations in tumors where RBSP3/HYA22 was expressed, growth suppression with regulated transgenes in culture, suppression of tumor formation in severe combined immunodeficient mice, and dephosphorylation of ppRB by RBSP3/HYA22, presumably leading to a block of the cell cycle at the G1/S boundary.


Subject(s)
Genes, Tumor Suppressor , Tumor Suppressor Proteins/genetics , Amino Acid Sequence , Base Sequence , Cell Division/genetics , Cell Line, Tumor , DNA Methylation , DNA Primers , DNA Probes , Gene Deletion , Humans , Microsatellite Repeats , Molecular Sequence Data , Phosphorylation , Polymerase Chain Reaction , RNA Splicing , Sequence Homology, Amino Acid , Sequence Tagged Sites , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...