Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Cancers (Basel) ; 13(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34885204

ABSTRACT

The MYC transcription factor regulates a vast number of genes and is implicated in many human malignancies. In some hematological malignancies, MYC is frequently subject to missense mutations that enhance its transformation activity. Here, we use a novel murine cell system to (i) characterize the transcriptional effects of progressively increasing MYC levels as normal primary B-cells transform to lymphoma cells and (ii) determine how this gene regulation program is modified by lymphoma-associated MYC mutations (T58A and T58I) that enhance its transformation activity. Unlike many previous studies, the cell system exploits primary B-cells that are transduced to allow regulated MYC expression under circumstances where apoptosis and senescence pathways are abrogated by the over-expression of the Bcl-xL and BMI1 proteins. In such cells, transition from a normal to a lymphoma phenotype is directly dependent on the MYC expression level, without a requirement for secondary events that are normally required during MYC-driven oncogenic transformation. A generalized linear model approach allowed an integrated analysis of RNA sequencing data to identify regulated genes in relation to both progressively increasing MYC level and wild type or mutant status. Using this design, a total of 7569 regulated genes were identified, of which the majority (n = 7263) were regulated in response to progressively increased levels of wild type MYC, while a smaller number of genes (n = 917) were differentially regulated, compared to wild type MYC, in T58A MYC- and/or T58I MYC-expressing cells. Unlike most genes that are similarly regulated by both wild type and mutant MYC genes, the set of 917 genes did not significantly overlap with known lipopolysaccharide regulated genes, which represent genes regulated by MYC in normal B cells. The genes that were differently regulated in cells expressing mutant MYC proteins were significantly enriched in DNA replication and G2 phase to mitosis transition genes. Thus, mutants affecting MYC proteins may augment quantitative oncogenic effects on the expression of normal MYC-target genes with qualitative oncogenic effects, by which sets of cell cycle genes are abnormally targeted by MYC as B cells transition into lymphoma cells. The T58A and T58I mutations augment MYC-driven transformation by distinct mechanisms.

2.
Cell Cycle ; 20(1): 23-38, 2021 01.
Article in English | MEDLINE | ID: mdl-33356836

ABSTRACT

Deregulated expression of the MYC oncogene is a frequent event during tumorigenesis and generally correlates with aggressive disease and poor prognosis. While MYC is a potent inducer of apoptosis, it often suppresses cellular senescence, which together with apoptosis is an important barrier against tumor development. For this latter function, MYC is dependent on cyclin-dependent kinase 2 (CDK2). Here, we utilized a MYC/BCL-XL-driven mouse model of acute myeloblastic leukemia (AML) to investigate whether pharmacological inhibition of CDK2 can inhibit MYC-driven tumorigenesis through induction of senescence. Purified mouse hematopoietic stem cells transduced with MYC and BCL-XL were transplanted into lethally irradiated mice, leading to the development of massive leukemia and subsequent death 15-17 days after transplantation. Upon disease onset, mice were treated with the selective CDK2 inhibitor CVT2584 or vehicle either by daily intraperitoneal injections or continuous delivery via mini-pumps. CVT2584 treatment delayed disease onset and moderately but significantly improved survival of mice. Flow cytometry revealed a significant decrease in tumor load in the spleen, liver and bone marrow of CVT2584-treated compared to vehicle-treated mice. This was correlated with induced senescence evidenced by reduced cell proliferation, increased senescence-associated ß-galactosidase activity and heterochromatin foci, expression of p19ARF and p21CIP1, and reduced phosphorylation (activation) of pRb, while very few apoptotic cells were observed. In addition, phosphorylation of MYC at Ser-62 was decreased. In summary, inhibition of CDK2 delayed MYC/BCL-XL-driven AML linked to senescence induction. Our results suggest that CDK2 is a promising target for pro-senescence cancer therapy, in particular for MYC-driven tumors, including leukemia.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Leukemia/metabolism , Proto-Oncogene Proteins c-myc/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Female , Humans , Leukemia/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation/genetics
3.
Eur J Immunol ; 49(3): 454-461, 2019 03.
Article in English | MEDLINE | ID: mdl-30664244

ABSTRACT

An experimental system where defined alterations in gene function or gene expression levels in primary B cells would result in the development of transformed plasma cells in vitro would be useful in order to facilitate studies of the underlying molecular mechanisms of plasma cell malignancies. Here, such a system is described in which primary murine B cells rapidly become transformed into surface CD138+ , IgM-/low , CD19- IgM-secreting plasma cells as a result of expression of the transcription factors IRF4 and MYC together with simultaneous expression of BMI1, mutated p53 or silencing of p19Arf , and suppression of intrinsic apoptosis through expression of BCLXL. Analysis of gene expression patterns revealed that this combination of transforming genes resulted in expression of a number of genes previously associated with terminally differentiated B cells (plasma cells) and myeloma cells, whereas many genes associated with mature B cells and B-cell lymphomas were not expressed. Upon transplantation, the transformed cells preferentially localized to the bone marrow, presenting features of a plasma cell malignancy of the IgM isotype. The present findings may also be applicable in the development of novel methods for production of monoclonal antibodies.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Transformation, Neoplastic/immunology , Lymphoma, B-Cell/immunology , Plasma Cells/immunology , Animals , Antigens, CD19/immunology , Antigens, CD19/metabolism , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Mice , Plasma Cells/metabolism , Plasma Cells/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-myc/metabolism
4.
Eur J Immunol ; 49(3): 375-385, 2019 03.
Article in English | MEDLINE | ID: mdl-30281155

ABSTRACT

Increased expression of the oncogene MYC is a common feature of many B-cell malignancies, however MYC overexpression by itself is not sufficient for transformation, and additional genetic events are required, although the exact nature of these remains unknown. In patients and in transgenic mouse models, oncogenic transformation may occur in B cells at various differentiation stages interacting with complex microenvironments. B-cell oncogenesis often occurs after prolonged periods of time, making it difficult to accurately identify the genetic events required for transformation. An in vitro system, where malignant transformation of primary B cells could be analyzed, would facilitate the identification of genetic events required for transformation. Here, we describe such a system and show that primary murine B cells rapidly become transformed upon forced expression of MYC, in conjunction with simultaneous inhibition of the ARF/p53 axis via overexpression of BMI1, as well as through downregulation of p19ARF or expression of a dominant-negative p53 and suppression of intrinsic apoptosis through overexpression of BCLXL or MCL1. Established tumor cells remained addicted to expression of the lymphoma-inducing genes. In mice, transformed cells rapidly established fatal B-cell lymphomas. Our results suggest that transformation of normal mature B cells into lymphomas can occur as a consequence of three defined events.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Cell Transformation, Neoplastic/immunology , Lymphoma, B-Cell/immunology , Proto-Oncogene Proteins c-myc/immunology , Tumor Suppressor Protein p53/immunology , Animals , Apoptosis/genetics , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression/immunology , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/immunology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Oncotarget ; 9(30): 21396-21410, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29765548

ABSTRACT

Peripheral T-cell lymphoma is an aggressive non-Hodgkin's lymphoma characterized by excessive proliferation of transformed mature T cells. The number and nature of genetic aberrations required and sufficient for transformation of normal T cells into lymphomas is unknown. Here, using a combinatorial in vitro-approach, we demonstrate that overexpression of MYC together with activated AKT in conditions of inhibition of intrinsic apoptosis rapidly resulted in transformation of mature mouse T cells with a frequency approaching 100%. Injection of transformed cells into mice resulted in rapid development of aggressive T cell lymphoma, characterized by spread to several organs, destruction of tissue architecture and rapid death of the animals. TcR-sequencing revealed a polyclonal repertoire of tumor cells indicating that co-expression of MYC, activated AKT and BCLXL is sufficient for tumor transformation and do not require acquisition of additional genetic events. When analyzing cells with inducible expression we found that proliferation of transformed T cells required sustained expression of both MYC and AKT. AKT exerted a dual function as it inhibited induction of, and promoted exit from, cellular quiescence and contributed to inhibion of apoptosis. Downregulation of AKT and/or MYC together with BCLXL resulted in rapid and complete elimination of cells through induction of apoptotic cell death.

7.
Haematologica ; 102(3): 498-508, 2017 03.
Article in English | MEDLINE | ID: mdl-27884971

ABSTRACT

A high proportion of patients with lower-risk del(5q) myelodysplastic syndromes will respond to treatment with lenalidomide. The median duration of transfusion-independence is 2 years with some long-lasting responses, but almost 40% of patients progress to acute leukemia by 5 years after starting treatment. The mechanisms underlying disease progression other than the well-established finding of small TP53-mutated subclones at diagnosis remain unclear. We studied a longitudinal cohort of 35 low- and intermediate-1-risk del(5q) patients treated with lenalidomide (n=22) or not (n=13) by flow cytometric surveillance of hematopoietic stem and progenitor cell subsets, targeted sequencing of mutational patterns, and changes in the bone marrow microenvironment. All 13 patients with disease progression were identified by a limited number of mutations in TP53, RUNX1, and TET2, respectively, with PTPN11 and SF3B1 occurring in one patient each. TP53 mutations were found in seven of nine patients who developed acute leukemia, and were documented to be present in the earliest sample (n=1) and acquired during lenalidomide treatment (n=6). By contrast, analysis of the microenvironment, and of hematopoietic stem and progenitor cells by flow cytometry was of limited prognostic value. Based on our data, we advocate conducting a prospective study aimed at investigating, in a larger number of cases of del(5q) myelodysplastic syndromes, whether the detection of such mutations before and after lenalidomide treatment can guide clinical decision-making.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 5 , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Aged , Aged, 80 and over , Biomarkers , Computational Biology/methods , Disease Progression , Female , Gene Expression , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Lenalidomide , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Myelodysplastic Syndromes/therapy , Prognosis , Stem Cell Niche , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Treatment Outcome
8.
Oncotarget ; 8(27): 43853-43865, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28029652

ABSTRACT

A putative tumor suppressor BLU mapped on the chromosomal 3p21 region, is frequently lost in human tumors including nasopharyngeal carcinoma (NPC). To explore the underlying mechanism of tumor suppression by BLU, its potential to promote apoptosis induced by TRAIL, an effector molecule elaborated by natural killer-T (NKT) cells was investigated. BLU was re-expressed in NPC-derived HNE1 cells by recombinant adenoviral infection and the cells were challenged with recombinant TRAIL. The growth inhibition of BLU was assayed and apoptosis was examined by flow cytometry-based tetramethylrhodamine ethyl ester (TMRE) and annexin V staining, cleavage of pro-caspase-8 and poly ADP ribose polymerase (PARP). The modulation of NF-κB pathway by BLU was evaluated by the reporter activity and estimation of the level of the molecules involved such as IKKalpha, p65 NF-κB, as well as NF-κB induced anti-apoptotic factors cFLIPL and cIAP2. The expression of BLU exerted in vitro and in vivo growth inhibitory effect and promoted TRAIL-induced apoptosis. This phenomenon was validated by FACS-based assays of mitochondrial membrane potential (BLU vs. Vector 87.8% ± 7.7% and 72.1%±6.7% at 6h exposure to TRAIL) and phosphatidylserine turnover (BLU vs. vector: 28.7%±2.9% and 22.6%±2.5%), as well as, enhanced caspapse-8 cleavage. Similar with the findings that BLU promotes chemotherapeutic agent-induced apoptosis, it also augmented death receptor-induced pathway through NF-κB pathway inhibition. In conclusion, BLU suppressed tumor formation by strengthening the antitumor immunity.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Carcinoma/genetics , Carcinoma/metabolism , NF-kappa B/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Suppressor Proteins/genetics , Animals , Carcinoma/immunology , Carcinoma/pathology , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival , Cytoskeletal Proteins , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Mice , Models, Biological , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Xenograft Model Antitumor Assays
11.
Oncotarget ; 6(41): 43679-97, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26544897

ABSTRACT

Despite recent advances in targeted therapeutics, administration of 5-fluorouracil (5-FU) remains a common clinical strategy for post-surgical treatment of solid tumors. Although it has been proposed that RNA metabolism is disturbed by 5-FU treatment, the key cytotoxic response is believed to be enzymatic inhibition of thymidylate synthase resulting in nucleotide pool disproportions. An operating p53 tumor suppressor signaling network is in many cases essential for the efficiency of chemotherapy, and malfunctions within this system remain a clinical obstacle. Since the fate of chemotherapy-insensitive tumor cells is rarely described, we performed a comparative analysis of 5-FU toxicity in p53-deficient cells and conclude that p53 acts as a facilitator rather than a gatekeeper of cell death. Although p53 can act as a regulator of several cellular stress responses, no rerouting of cell death mode was observed in absence of the tumor suppressor. Thus, the final death outcome of 5-FU-treated p53-/- cells is demonstrated to be caspase-dependent, but due to a slow pace, accumulation of mitochondrial reactive oxygen species contributes to necrotic characteristics. The oligomerization status of the p53 target gene DR5 is determined as a significant limiting factor for the initiation of caspase activity in an intracellular TRAIL-dependent manner. Using several experimental approaches, we further conclude that RNA-rather than DNA-related stress follows by caspase activation irrespectively of p53 status. A distinct 5-FU-induced stress mechanism is thereby functionally connected to a successive and discrete cell death signaling pathway. Finally, we provide evidence that silencing of PARP-1 function may be an approach to specifically target p53-deficient cells in 5-FU combinatorial treatment strategies. Together, our results disclose details of impaired cell death signaling engaged as a consequence of 5-FU chemotherapy. Obtained data will contribute to the comprehension of factors restraining 5-FU efficiency, and by excluding DNA as the main stress target in some cell types they propose alternatives to currently used and suggested synergistic treatment regimens.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Fluorouracil/pharmacology , RNA/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Electron, Transmission , Polymerase Chain Reaction , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transduction, Genetic , Tumor Suppressor Protein p53/metabolism
12.
J Leukoc Biol ; 96(3): 481-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24899587

ABSTRACT

This manuscript describes technical advances allowing manipulation and quantitative analyses of human DC migratory behavior in lung epithelial tissue. DCs are hematopoietic cells essential for the maintenance of tissue homeostasis and the induction of tissue-specific immune responses. Important functions include cytokine production and migration in response to infection for the induction of proper immune responses. To design appropriate strategies to exploit human DC functional properties in lung tissue for the purpose of clinical evaluation, e.g., candidate vaccination and immunotherapy strategies, we have developed a live-imaging assay based on our previously described organotypic model of the human lung. This assay allows provocations and subsequent quantitative investigations of DC functional properties under conditions mimicking morphological and functional features of the in vivo parental tissue. We present protocols to set up and prepare tissue models for 4D (x, y, z, time) fluorescence-imaging analysis that allow spatial and temporal studies of human DCs in live epithelial tissue, followed by flow cytometry analysis of DCs retrieved from digested tissue models. This model system can be useful for elucidating incompletely defined pathways controlling DC functional responses to infection and inflammation in lung epithelial tissue, as well as the efficacy of locally administered candidate interventions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cell Culture Techniques , Dendritic Cells/drug effects , Lung/immunology , Time-Lapse Imaging/methods , Cell Communication , Cell Line , Cell Movement , Chemokine CCL2/pharmacology , Coculture Techniques , Culture Media, Conditioned , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/cytology , Dendritic Cells/immunology , Epithelial Cells/cytology , Fibroblasts/cytology , Flow Cytometry , Gene Expression Profiling , Genes, Reporter , Humans , Inflammation/chemically induced , Inflammation/immunology , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Lung/cytology , Models, Immunological , Monocytes/cytology , Recombinant Proteins/pharmacology
13.
FEBS Lett ; 588(17): 2921-7, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24910348

ABSTRACT

HS-1-associated protein X-1 (HAX-1) is a multi-functional protein that has been implicated in the regulation of apoptosis, cell motility and calcium homeostasis. In the present study, we set out to assess the postulated functional resemblance of HAX-1 to the BCL-2 family of anti-apoptotic proteins using non-transformed, cytokine-dependent murine bone marrow cells as a model system. BCL-X(L), but not HAX-1 protected against cytokine withdrawal-induced apoptosis while HAX-1 and BCL-X(L) significantly reduced thapsigargin-triggered (calcium-dependent) apoptosis. The data argue in favor of cell type- and stimulus-specific roles of HAX-1 in regulation of cell survival.


Subject(s)
Apoptosis , Bone Marrow Cells/cytology , Cytokines/metabolism , Proteins/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Thapsigargin/pharmacology
14.
PLoS One ; 8(6): e65936, 2013.
Article in English | MEDLINE | ID: mdl-23840379

ABSTRACT

Acetylcholine (ACh), the classical neurotransmitter, also affects a variety of nonexcitable cells, such as endothelia, microglia, astrocytes and lymphocytes in both the nervous system and secondary lymphoid organs. Most of these cells are very distant from cholinergic synapses. The action of ACh on these distant cells is unlikely to occur through diffusion, given that ACh is very short-lived in the presence of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), two extremely efficient ACh-degrading enzymes abundantly present in extracellular fluids. In this study, we show compelling evidence for presence of a high concentration and activity of the ACh-synthesizing enzyme, choline-acetyltransferase (ChAT) in human cerebrospinal fluid (CSF) and plasma. We show that ChAT levels are physiologically balanced to the levels of its counteracting enzymes, AChE and BuChE in the human plasma and CSF. Equilibrium analyses show that soluble ChAT maintains a steady-state ACh level in the presence of physiological levels of fully active ACh-degrading enzymes. We show that ChAT is secreted by cultured human-brain astrocytes, and that activated spleen lymphocytes release ChAT itself rather than ACh. We further report differential CSF levels of ChAT in relation to Alzheimer's disease risk genotypes, as well as in patients with multiple sclerosis, a chronic neuroinflammatory disease, compared to controls. Interestingly, soluble CSF ChAT levels show strong correlation with soluble complement factor levels, supporting a role in inflammatory regulation. This study provides a plausible explanation for the long-distance action of ACh through continuous renewal of ACh in extracellular fluids by the soluble ChAT and thereby maintenance of steady-state equilibrium between hydrolysis and synthesis of this ubiquitous cholinergic signal substance in the brain and peripheral compartments. These findings may have important implications for the role of cholinergic signaling in states of inflammation in general and in neurodegenerative disease, such as Alzheimer's disease and multiple sclerosis in particular.


Subject(s)
Alzheimer Disease/enzymology , Astrocytes/enzymology , Choline O-Acetyltransferase/blood , Choline O-Acetyltransferase/cerebrospinal fluid , Multiple Sclerosis/enzymology , Acetylcholine/metabolism , Acetylcholinesterase/blood , Acetylcholinesterase/cerebrospinal fluid , Alzheimer Disease/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Butyrylcholinesterase/blood , Butyrylcholinesterase/cerebrospinal fluid , Cells, Cultured , Choline O-Acetyltransferase/genetics , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Genotype , Humans , Lymphocytes/cytology , Lymphocytes/enzymology , Mice , Multiple Sclerosis/genetics
15.
Leukemia ; 27(4): 889-896, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23070040

ABSTRACT

Refractory anemia with ring sideroblasts (RARS) is characterized by mitochondrial ferritin (FTMT) accumulation and markedly suppressed expression of the iron transporter ABCB7. To test the hypothesis that ABCB7 is a key mediator of ineffective erythropoiesis of RARS, we modulated its expression in hematopoietic cells. ABCB7 up and downregulation did not influence growth and survival of K562 cells. In normal bone marrow, ABCB7 downregulation reduced erythroid differentiation, growth and colony formation, and resulted in a gene expression pattern similar to that observed in intermediate RARS erythroblasts, and in the accumulation of FTMT. Importantly, forced ABCB7 expression restored erythroid colony growth and decreased FTMT expression level in RARS CD34+ marrow cells. Mutations in the SF3B1 gene, a core component of the RNA splicing machinery, were recently identified in a high proportion of patients with RARS and 11 of the 13 RARS patients in this study carried this mutation. Interestingly, ABCB7 exon usage differed between normal bone marrow and RARS, as well as within the RARS cohort. In addition, SF3B1 silencing resulted in downregulation of ABCB7 in K562 cells undergoing erythroid differentiation. Our findings support that ABCB7 is implicated in the phenotype of acquired RARS and suggest a relation between SF3B1 mutations and ABCB7 downregulation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Anemia, Refractory/genetics , Anemia, Sideroblastic/genetics , Aged , Aged, 80 and over , Cohort Studies , Down-Regulation , Exons , Female , Flow Cytometry , Gene Silencing , Humans , Immunohistochemistry , K562 Cells , Male , Middle Aged , Phenotype , RNA Splicing , Real-Time Polymerase Chain Reaction
16.
EMBO J ; 32(1): 86-99, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23222484

ABSTRACT

Infection of macrophages by bacterial pathogens can trigger Toll-like receptor (TLR) activation as well as Nod-like receptors (NLRs) leading to inflammasome formation and cell death dependent on caspase-1 (pyroptosis). Complicating the study of inflammasome activation is priming. Here, we develop a priming-free NLRC4 inflammasome activation system to address the necessity and role of priming in pyroptotic cell death and damage-associated molecular pattern (DAMP) release. We find pyroptosis is not dependent on priming and when priming is re-introduced pyroptosis is unaffected. Cells undergoing unprimed pyroptosis appear to be independent of mitochondrial involvement and do not produce inflammatory cytokines, nitrous oxide (NO), or reactive oxygen species (ROS). Nevertheless, they undergo an explosive cell death releasing a chemotactic isoform of the DAMP high mobility group protein box 1 (HMGB1). Importantly, priming through surface TLRs but not endosomal TLRs during pyroptosis leads to the release of a new TLR4-agonist cysteine redox isoform of HMGB1. These results show that pyroptosis is dominant to priming signals and indicates that metabolic changes triggered by priming can affect how cell death is perceived by the immune system.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , HMGB1 Protein/metabolism , Macrophages/immunology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Toll-Like Receptors/metabolism , Acetylation , Amino Acid Sequence , Animals , Apoptosis , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/immunology , Bacterial Proteins/metabolism , Calcium-Binding Proteins/agonists , Calcium-Binding Proteins/immunology , Cell Death , Cell Line , Gene Expression , HMGB1 Protein/analysis , Host-Pathogen Interactions , Inflammasomes/immunology , Inflammasomes/metabolism , Macrophage Activation/physiology , Macrophages/microbiology , Macrophages/physiology , Mice , Molecular Sequence Data , Neuronal Apoptosis-Inhibitory Protein/agonists , Neuronal Apoptosis-Inhibitory Protein/immunology , Protein Isoforms/metabolism , Signal Transduction , Toll-Like Receptors/immunology
17.
PLoS One ; 7(2): e31366, 2012.
Article in English | MEDLINE | ID: mdl-22393362

ABSTRACT

Myc plays an important role in tumor development, including acute myeloid leukemia (AML). However, MYC is also a powerful inducer of apoptosis, which is one of the major failsafe programs to prevent cancer development. To clarify the relative importance of the extrinsic (death receptor-mediated) versus the intrinsic (mitochondrial) pathway of apoptosis in MYC-driven AML, we coexpressed MYC together with anti-apoptotic proteins of relevance for AML; BCL-X(L)/BCL-2 (inhibiting the intrinsic pathway) or FLIP(L) (inhibiting the extrinsic pathway), in hematopoietic stems cells (HSCs). Transplantation of HSCs expressing MYC into syngeneic recipient mice resulted in development of AML and T-cell lymphomas within 7-9 weeks as expected. Importantly, coexpression of MYC together with BCL-X(L)/BCL-2 resulted in strongly accelerated kinetics and favored tumor development towards aggressive AML. In contrast, coexpression of MYC and FLIP(L) did neither accelerate tumorigenesis nor change the ratio of AML versus T-cell lymphoma. However, a change in distribution of immature CD4(+)CD8(+) versus mature CD4(+) T-cell lymphoma was observed in MYC/FLIP(L) mice, possibly as a result of increased survival of the CD4+ population, but this did not significantly affect the outcome of the disease. In conclusion, our findings provide direct evidence that BCL-X(L) and BCL-2 but not FLIP(L) acts in synergy with MYC to drive AML development.


Subject(s)
Apoptosis , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Bacterial Proteins/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Humans , Kinetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Models, Biological , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
19.
PLoS One ; 5(11): e14085, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21124928

ABSTRACT

The SRY (sex determining region Y)-box 11 (SOX11) gene, located on chromosome 2p25, encodes for a transcription factor that is involved in tissue remodeling during embryogenesis and is crucial for neurogenesis. The role for SOX11 in hematopoiesis has not yet been defined. Two genes under direct control of SOX11 are the class- III ß-tubulin gene (TUBB3) in neural cells and the transcription factor TEA domain family member 2 (TEAD2) in neural and mesenchymal progenitor cells. Normal, mature lymphocytes lack SOX11 but express SOX4, another member of the same group of SOX transcription factors. We and others recently identified SOX11 as aberrantly expressed in mantle cell lymphoma (MCL). Since SOX11 is variably expressed in MCL it may not be essential for tumorigenesis, but may carry prognostic information. Currently, no specific functional effects have been linked to SOX11 expression in MCL and it is not known which genes are under influence of SOX11 in lymphoma. In this study we found variable expression of SOX11, SOX4 and SOX12 mRNA in mantle cell lymphoma cell lines. Downregulation of SOX11 expression by siRNA verified that SOX11 controlled the expression of the gene TUBB3 in the MCL cell line Granta 519. Furthermore we identified, by global gene expression analysis, 26 new target genes influenced by siRNA SOX11 downmodulation. Among these genes, DBN1, SETMAR and HIG2 were found to be significantly correlated to SOX11 expression in two cohorts of primary mantle cell lymphomas. Chromatin immunoprecipitation (ChIP) analysis showed that these genes are direct targets of the SOX11 protein. In spite of almost complete downregulation of the SOX11 protein no significant effects on Granta 519 cell proliferation or survival in short term in vitro experiments was found. In summary we have identified a number of genes influenced by SOX11 expression in MCL cell lines and primary MCL. Among these genes, DBN1, SETMAR and HIG2 are direct transcriptional targets of the SOX11 protein.


Subject(s)
Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Lymphoma, Mantle-Cell/genetics , Neoplasm Proteins/genetics , Neuropeptides/genetics , SOXC Transcription Factors/genetics , Aged , Aged, 80 and over , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Male , Neoplasm Proteins/metabolism , Neuropeptides/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , SOXC Transcription Factors/metabolism , Tubulin/genetics , Tubulin/metabolism
20.
J Inherit Metab Dis ; 33(3): 231-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20440651

ABSTRACT

Mitochondria are important for normal blood-cell development, and several diseases linked to mitochondrial DNA (mtDNA) show hematological manifestations. We recently generated a mouse strain deficient in expression of the mitochondrial pyrimidine nucleoside kinase thymidine kinase 2 (Tk2), showing that these mice exhibit progressive mtDNA depletion in multiple organs. We used this mouse strain as a model for mtDNA depletion syndromes to investigate the effects of mtDNA depletion on hematopoiesis. MtDNA levels in spleen from the Tk2-deficient mice were decreased 50%, but in contrast to all other investigated organs, both thymus and peripheral blood leukocytes showed normal mtDNA levels. Analysis of peripheral blood and cell populations in spleen, thymus, and bone marrow showed normal findings in the Tk2-deficient mice. The total rates of thymidine phosphorylation-which also include phosphorylation catalyzed by cytosolic Tk 1-in both spleen and thymus from wild-type mice were >50-fold higher than in liver, brain, and muscle. In summary, our data show that blood cells are less dependent on mitochondrial Tk2 compared with several other tissues and that these cells can synthesize deoxyribonucleotides required for mtDNA replication by alternative pathways such as phosphorylation of thymidine by cytosolic Tk1.


Subject(s)
DNA, Mitochondrial/metabolism , Hematopoiesis , Thymidine Kinase/genetics , Animals , Cell Proliferation , Disease Models, Animal , Embryonic Stem Cells/cytology , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...