Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 162(4): 727-37, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276629

ABSTRACT

Chronic infection with Plasmodium falciparum was epidemiologically associated with endemic Burkitt's lymphoma, a mature B cell cancer characterized by chromosome translocation between the c-myc oncogene and Igh, over 50 years ago. Whether infection promotes B cell lymphoma, and if so by which mechanism, remains unknown. To investigate the relationship between parasitic disease and lymphomagenesis, we used Plasmodium chabaudi (Pc) to produce chronic malaria infection in mice. Pc induces prolonged expansion of germinal centers (GCs), unique compartments in which B cells undergo rapid clonal expansion and express activation-induced cytidine deaminase (AID), a DNA mutator. GC B cells elicited during Pc infection suffer widespread DNA damage, leading to chromosome translocations. Although infection does not change the overall rate, it modifies lymphomagenesis to favor mature B cell lymphomas that are AID dependent and show chromosome translocations. Thus, malaria infection favors mature B cell cancers by eliciting protracted AID expression in GC B cells. PAPERCLIP.


Subject(s)
Genomic Instability , Lymphoma, B-Cell/genetics , Malaria/complications , Malaria/genetics , Plasmodium chabaudi/physiology , Animals , B-Lymphocytes/pathology , Chronic Disease , Cytidine Deaminase/metabolism , DNA Replication , Genes, p53 , Germinal Center/parasitology , Malaria/parasitology , Malaria/pathology , Mice , Translocation, Genetic
2.
Cell ; 141(2): 243-54, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20362325

ABSTRACT

Defective DNA repair by homologous recombination (HR) is thought to be a major contributor to tumorigenesis in individuals carrying Brca1 mutations. Here, we show that DNA breaks in Brca1-deficient cells are aberrantly joined into complex chromosome rearrangements by a process dependent on the nonhomologous end-joining (NHEJ) factors 53BP1 and DNA ligase 4. Loss of 53BP1 alleviates hypersensitivity of Brca1 mutant cells to PARP inhibition and restores error-free repair by HR. Mechanistically, 53BP1 deletion promotes ATM-dependent processing of broken DNA ends to produce recombinogenic single-stranded DNA competent for HR. In contrast, Lig4 deficiency does not rescue the HR defect in Brca1 mutant cells but prevents the joining of chromatid breaks into chromosome rearrangements. Our results illustrate that HR and NHEJ compete to process DNA breaks that arise during DNA replication and that shifting the balance between these pathways can be exploited to selectively protect or kill cells harboring Brca1 mutations.


Subject(s)
BRCA1 Protein/genetics , DNA Repair , Intracellular Signaling Peptides and Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Chromosomal Proteins, Non-Histone , DNA Breaks , DNA-Binding Proteins , Female , Genomic Instability , Humans , Mice , Tumor Suppressor p53-Binding Protein 1
3.
Br J Haematol ; 204(3): 945-958, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38296260

ABSTRACT

EVI1 expression is associated with poor prognosis in myeloid leukaemia, which can result from Chr.3q alterations that juxtapose enhancers to induce EVI1 expression via long-range chromatin interactions. More often, however, EVI1 expression occurs unrelated to 3q alterations, and it remained unclear if, in these cases, EVI1 expression is similarly caused by aberrant enhancer activation. Here, we report that, in EVI1+3q- myeloid leukaemia cells, the EVI1 promoter interacts via long-range chromatin interactions with promoters of distally located, active genes, rather than with enhancer elements. Unlike in 3q+ cells, EVI1 expression and long-range interactions appear to not depend on CTCF/cohesin, though EVI1+3q- cells utilise an EVI1 promoter-proximal site to enhance its expression that is also involved in CTCF-mediated looping in 3q+ cells. Long-range interactions in 3q- cells connect EVI1 to promoters of multiple genes, whose transcription correlates with EVI1 in EVI1+3q- cell lines, suggesting a shared mechanism of transcriptional regulation. In line with this, CRISPR interference-induced silencing of two of these sites minimally, but consistently reduced EVI1 expression. Together, we provide novel evidence of features associated with EVI1 expression in 3q- leukaemia and consolidate the view that EVI1 in 3q- leukaemia is largely promoter-driven, potentially involving long-distance promoter clustering.


Subject(s)
Leukemia, Myeloid , Transcription Factors , Humans , Transcription Factors/genetics , DNA-Binding Proteins/genetics , Chromatin , MDS1 and EVI1 Complex Locus Protein/genetics , Leukemia, Myeloid/genetics , Proto-Oncogenes
4.
J Immunol ; 202(12): 3423-3433, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31085591

ABSTRACT

Induction of programmed DNA damage and its recognition and repair are fundamental for B cell development. The ssDNA-binding protein SSB1 has been described in human cells as essential for the recognition and repair of DNA damage. To study its relevance for B cells, we recently developed Ssb1 -/- and conditional Ssb1 -/- mice. Although SSB1 loss did not affect B cell development, Ssb1 -/- cells exhibited compensatory expression of its homolog SSB2. We have now generated Ssb2 -/- mice and show in this study that SSB2 is also dispensable for B cell development and DNA damage response activation. In contrast to the single loss of Ssb1 or Ssb2, however, combined SSB1/2 deficiency caused a defect in early B cell development. We relate this to the sensitivity of B cell precursors as mature B cells largely tolerated their loss. Toxicity of combined genetic SSB1/2 loss can be rescued by ectopic expression of either SSB1 or SSB2, mimicked by expression of SSB1 ssDNA-binding mutants, and attenuated by BCL2-mediated suppression of apoptosis. SSB1/2 loss in B cell precursors further caused increased exposure of ssDNA associated with disruption of genome fragile sites, inefficient cell cycle progression, and increased DNA damage if apoptosis is suppressed. As such, our results establish SSB1/2 as safeguards of B cell development and unveil their differential requirement in immature and mature B lymphocytes.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/metabolism , Precursor Cells, B-Lymphoid/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Apoptosis , Cell Differentiation , Cells, Cultured , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Genome/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Suppressor of Cytokine Signaling Proteins/genetics
5.
Mol Cell ; 49(4): 623-31, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23290917

ABSTRACT

Deficiencies in factors that regulate the DNA damage response enhance the incidence of malignancy by destabilizing the genome. However, the precise influence of the DNA damage response on regulation of cancer-associated rearrangements is not well defined. Here we examine the genome-wide impact of tumor protein P53-binding protein 1 (53BP1) deficiency in lymphoma and translocation. While both activation-induced cytidine deaminase (AID) and 53BP1 have been associated with cancer in humans, neither AID overexpression nor loss of 53BP1 is sufficient to produce malignancy. However, the combination of 53BP1 deficiency and AID deregulation results in B cell lymphoma. Deep sequencing of the genome of 53BP1(-/-) cancer cells and translocation capture sequencing (TC-Seq) of primary 53BP1(-/-) B cells revealed that their chromosomal rearrangements differ from those found in wild-type cells in that they show increased DNA end resection. Moreover, loss of 53BP1 alters the translocatome by increasing rearrangements to intergenic regions.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Proteins, Non-Histone/physiology , Cytidine Deaminase/physiology , DNA-Binding Proteins/physiology , Gene Rearrangement , Lymphoma, B-Cell/metabolism , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Mammalian/genetics , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Genes, Tumor Suppressor , Genome-Wide Association Study , Lymphoma, B-Cell/genetics , Mice , Mice, Knockout , Mutation , Sequence Analysis, DNA , Transcription, Genetic , Translocation, Genetic , Tumor Suppressor p53-Binding Protein 1
7.
Mol Cell ; 42(3): 319-29, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21549309

ABSTRACT

53BP1 is a DNA damage protein that forms phosphorylated H2AX (γ-H2AX) dependent foci in a 1 Mb region surrounding DNA double-strand breaks (DSBs). In addition, 53BP1 promotes genomic stability by regulating the metabolism of DNA ends. We have compared the joining rates of paired DSBs separated by 1.2 kb to 27 Mb on chromosome 12 in the presence or absence of 53BP1. 53BP1 facilitates joining of intrachromosomal DSBs but only at distances corresponding to γ-H2AX spreading. In contrast, DNA end protection by 53BP1 is distance independent. Furthermore, analysis of 53BP1 mutants shows that chromatin association, oligomerization, and N-terminal ATM phosphorylation are all required for DNA end protection and joining as measured by immunoglobulin class switch recombination. These data elucidate the molecular events that are required for 53BP1 to maintain genomic stability and point to a model wherein 53BP1 and H2AX cooperate to repress resection of DSBs.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Immunoglobulin Class Switching/genetics , Animals , B-Lymphocytes/metabolism , Binding Sites , Blotting, Western , Cells, Cultured , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Flow Cytometry , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Phosphorylation , Protein Multimerization , Recombination, Genetic , Tumor Suppressor p53-Binding Protein 1
8.
Mol Cell ; 36(4): 631-41, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19941823

ABSTRACT

Cancer-initiating translocations such as those associated with lymphomas require the formation of paired DNA double-strand breaks (DSBs). Activation-induced cytidine deaminase (AID) produces widespread somatic mutation in mature B cells; however, the extent of "off-target" DSB formation and its role in translocation-associated malignancy is unknown. Here, we show that deregulated expression of AID causes widespread genome instability, which alone is insufficient to induce B cell lymphoma; transformation requires concomitant loss of the tumor suppressor p53. Mature B cell lymphomas arising as a result of deregulated AID expression are phenotypically diverse and harbor clonal reciprocal translocations involving a group of Immunoglobulin (Ig) and non-Ig genes that are direct targets of AID. This group includes miR-142, a previously unknown micro-RNA target that is translocated in human B cell malignancy. We conclude that AID produces DSBs throughout the genome, which can lead to lymphoma-associated chromosome translocations in mature B cells.


Subject(s)
Chromosomes, Mammalian/genetics , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , Genes, Immunoglobulin/genetics , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Translocation, Genetic , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Cell Differentiation/genetics , Cells, Cultured , Chromosomal Instability/genetics , DNA Damage , Humans , Immunoglobulin Class Switching/genetics , Karyotyping , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , MicroRNAs/metabolism , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Tumor Suppressor Protein p53/deficiency
9.
EMBO J ; 31(20): 4045-56, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22940690

ABSTRACT

Human single-stranded DNA-binding protein 1 (hSSB1), encoded by OBFC2B, was recently characterized as an essential factor for the initiation of DNA damage checkpoints and the maintenance of genomic stability. Here, we report that loss of Obfc2b in mice results in perinatal lethality characterized by growth delay and skeletal abnormalities. These abnormalities are associated with accumulation of γH2ax, apoptosis and defective pre-cartilage condensation, which is essential for normal bone formation. However, deficiency of Obfc2b does not affect the initiation of DNA damage checkpoints, Atm activation, or the maintenance of genomic stability in B lymphocytes and primary fibroblasts. Loss of Obfc2b results in increased expression of its homologue Obfc2a (hSSB2). In contrast to Obfc2b deficiency, depletion of Obfc2a in fibroblasts results in impaired proliferation, accumulation of γH2ax and increased genomic instability. Thus, the hSSB1 orthologue Obfc2b has a unique function during embryogenesis limited to cell types that contribute to bone formation. While being dispensable in most other cell lineages, its absence leads to a compensatory increase in Obfc2a protein, a homologue required for the maintenance of genomic integrity.


Subject(s)
DNA Damage , DNA Repair/genetics , DNA-Binding Proteins/deficiency , Mitochondrial Proteins/deficiency , Osteogenesis/genetics , Animals , Apoptosis/genetics , B-Lymphocytes/metabolism , Bone and Bones/abnormalities , Bone and Bones/embryology , Cell Division , Chondrocytes/metabolism , DNA Replication , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genes, Lethal , Genes, p53 , Histones/metabolism , Humans , Mesoderm/abnormalities , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Organ Specificity , Osteoblasts/metabolism , Osteoclasts/metabolism
10.
Hum Mutat ; 36(2): 187-90, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504470

ABSTRACT

Cerebro-costo-mandibular syndrome (CCMS) is a developmental disorder characterized by the association of Pierre Robin sequence and posterior rib defects. Exome sequencing and Sanger sequencing in five unrelated CCMS patients revealed five heterozygous variants in the small nuclear ribonucleoprotein polypeptides B and B1 (SNRPB) gene. This gene includes three transcripts, namely transcripts 1 and 2, encoding components of the core spliceosomal machinery (SmB' and SmB) and transcript 3 undergoing nonsense-mediated mRNA decay. All variants were located in the premature termination codon (PTC)-introducing alternative exon of transcript 3. Quantitative RT-PCR analysis revealed a significant increase in transcript 3 levels in leukocytes of CCMS individuals compared to controls. We conclude that CCMS is due to heterozygous mutations in SNRPB, enhancing inclusion of a SNRPB PTC-introducing alternative exon, and show that this developmental disease is caused by defects in the splicing machinery. Our finding confirms the report of SNRPB mutations in CCMS patients by Lynch et al. (2014) and further extends the clinical and molecular observations.


Subject(s)
Intellectual Disability/genetics , Micrognathism/genetics , Ribs/abnormalities , snRNP Core Proteins/genetics , Adolescent , Adult , Base Sequence , Child, Preschool , Genetic Association Studies , Heterozygote , Humans , Male , Mutation, Missense , Young Adult
11.
Nature ; 458(7238): 636-40, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19287373

ABSTRACT

Antibodies to conserved epitopes on the human immunodeficiency virus (HIV) surface protein gp140 can protect against infection in non-human primates, and some infected individuals show high titres of broadly neutralizing immunoglobulin (Ig)G antibodies in their serum. However, little is known about the specificity and activity of these antibodies. To characterize the memory antibody responses to HIV, we cloned 502 antibodies from HIV envelope-binding memory B cells from six HIV-infected patients with broadly neutralizing antibodies and low to intermediate viral loads. We show that in these patients, the B-cell memory response to gp140 is composed of up to 50 independent clones expressing high affinity neutralizing antibodies to the gp120 variable loops, the CD4-binding site, the co-receptor-binding site, and to a new neutralizing epitope that is in the same region of gp120 as the CD4-binding site. Thus, the IgG memory B-cell compartment in the selected group of patients with broad serum neutralizing activity to HIV is comprised of multiple clonal responses with neutralizing activity directed against several epitopes on gp120.


Subject(s)
B-Lymphocytes/immunology , HIV Antibodies/analysis , HIV Antibodies/immunology , HIV Infections/immunology , Immunologic Memory/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Antibody Affinity , Binding Sites , CD4 Antigens/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , HIV Antibodies/isolation & purification , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/immunology , Humans , Neutralization Tests , Receptors, HIV/metabolism , Viral Load , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/metabolism
12.
J Exp Med ; 204(5): 1157-66, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17485517

ABSTRACT

The Philadelphia chromosome (Ph) encoding the oncogenic BCR-ABL1 kinase defines a subset of acute lymphoblastic leukemia (ALL) with a particularly unfavorable prognosis. ALL cells are derived from B cell precursors in most cases and typically carry rearranged immunoglobulin heavy chain (IGH) variable (V) region genes devoid of somatic mutations. Somatic hypermutation is restricted to mature germinal center B cells and depends on activation-induced cytidine deaminase (AID). Studying AID expression in 108 cases of ALL, we detected AID mRNA in 24 of 28 Ph(+) ALLs as compared with 6 of 80 Ph(-) ALLs. Forced expression of BCR-ABL1 in Ph(-) ALL cells and inhibition of the BCR-ABL1 kinase showed that aberrant expression of AID depends on BCR-ABL1 kinase activity. Consistent with aberrant AID expression in Ph(+) ALL, IGH V region genes and BCL6 were mutated in many Ph(+) but unmutated in most Ph(-) cases. In addition, AID introduced DNA single-strand breaks within the tumor suppressor gene CDKN2B in Ph(+) ALL cells, which was sensitive to BCR-ABL1 kinase inhibition and silencing of AID expression by RNA interference. These findings identify AID as a BCR-ABL1-induced mutator in Ph(+) ALL cells, which may be relevant with respect to the particularly unfavorable prognosis of this leukemia subset.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/metabolism , Gene Expression Regulation, Neoplastic/immunology , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Protein-Tyrosine Kinases/genetics , B-Lymphocytes/metabolism , Base Sequence , Blotting, Western , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Flow Cytometry , Fusion Proteins, bcr-abl , Genes, myc/genetics , Humans , Immunoglobulin Variable Region/genetics , Molecular Sequence Data , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Oligonucleotides , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-6 , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment
13.
Blood Adv ; 7(21): 6395-6410, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37224458

ABSTRACT

Deregulated expression of lineage-affiliated transcription factors (TFs) is a major mechanism of oncogenesis. However, how the deregulation of nonlineage affiliated TF affects chromatin to initiate oncogenic transcriptional programs is not well-known. To address this, we studied the chromatin effects imposed by oncogenic MAF as the cancer-initiating driver in the plasma cell cancer multiple myeloma. We found that the ectopically expressed MAF endows myeloma plasma cells with migratory and proliferative transcriptional potential. This potential is regulated by the activation of enhancers and superenhancers, previously inactive in healthy B cells and plasma cells, and the cooperation of MAF with the plasma cell-defining TF IRF4. Forced ectopic MAF expression confirms the de novo ability of oncogenic MAF to convert transcriptionally inert chromatin to active chromatin with the features of superenhancers, leading to the activation of the MAF-specific oncogenic transcriptome and the acquisition of cancer-related cellular phenotypes such as CCR1-dependent cell migration. These findings establish oncogenic MAF as a pioneer transcription factor that can initiate as well as sustain oncogenic transcriptomes and cancer phenotypes. However, despite its pioneer function, myeloma cells remain MAF-dependent, thus validating oncogenic MAF as a therapeutic target that would be able to circumvent the challenges of subsequent genetic diversification driving disease relapse and drug resistance.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Gene Expression Regulation , Plasma Cells/metabolism , B-Lymphocytes/metabolism , Chromatin
14.
bioRxiv ; 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36993619

ABSTRACT

In most cell types, nuclear ß-catenin functions as prominent oncogenic driver and pairs with TCF7-family factors for transcriptional activation of MYC. Surprisingly, B-lymphoid malignancies not only lacked expression and activating lesions of ß-catenin but critically depended on GSK3ß for effective ß-catenin degradation. Our interactome studies in B-lymphoid tumors revealed that ß-catenin formed repressive complexes with lymphoid-specific Ikaros factors at the expense of TCF7. Instead of MYC-activation, ß-catenin was essential to enable Ikaros-mediated recruitment of nucleosome remodeling and deacetylation (NuRD) complexes for transcriptional repression of MYC. To leverage this previously unrecognized vulnerability of B-cell-specific repressive ß-catenin-Ikaros-complexes in refractory B-cell malignancies, we examined GSK3ß small molecule inhibitors to subvert ß-catenin degradation. Clinically approved GSK3ß-inhibitors that achieved favorable safety prof les at micromolar concentrations in clinical trials for neurological disorders and solid tumors were effective at low nanomolar concentrations in B-cell malignancies, induced massive accumulation of ß-catenin, repression of MYC and acute cell death. Preclinical in vivo treatment experiments in patient-derived xenografts validated small molecule GSK3ß-inhibitors for targeted engagement of lymphoid-specific ß-catenin-Ikaros complexes as a novel strategy to overcome conventional mechanisms of drug-resistance in refractory malignancies. HIGHLIGHTS: Unlike other cell lineages, B-cells express nuclear ß-catenin protein at low baseline levels and depend on GSK3ß for its degradation.In B-cells, ß-catenin forms unique complexes with lymphoid-specific Ikaros factors and is required for Ikaros-mediated tumor suppression and assembly of repressive NuRD complexes. CRISPR-based knockin mutation of a single Ikaros-binding motif in a lymphoid MYC superenhancer region reversed ß-catenin-dependent Myc repression and induction of cell death. The discovery of GSK3ß-dependent degradation of ß-catenin as unique B-lymphoid vulnerability provides a rationale to repurpose clinically approved GSK3ß-inhibitors for the treatment of refractory B-cell malignancies. GRAPHICAL ABSTRACT: Abundant nuclear ß-cateninß-catenin pairs with TCF7 factors for transcriptional activation of MYCB-cells rely on efficient degradation of ß-catenin by GSK3ßB-cell-specific expression of Ikaros factors Unique vulnerability in B-cell tumors: GSK3ß-inhibitors induce nuclear accumulation of ß-catenin.ß-catenin pairs with B-cell-specific Ikaros factors for transcriptional repression of MYC.

15.
BMC Genomics ; 13: 418, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22916712

ABSTRACT

BACKGROUND: Acute lymphoblastic leukemia (ALL) cells treated with drugs can become drug-tolerant if co-cultured with protective stromal mouse embryonic fibroblasts (MEFs). RESULTS: We performed transcriptional profiling on these stromal fibroblasts to investigate if they were affected by the presence of drug-treated ALL cells. These mitotically inactivated MEFs showed few changes in gene expression, but a family of sequences of which transcription is significantly increased was identified. A sequence related to this family, which we named cassini, was selected for further characterization. We found that cassini was highly upregulated in drug-treated ALL cells. Analysis of RNAs from different normal mouse tissues showed that cassini expression is highest in spleen and thymus, and can be further enhanced in these organs by exposure of mice to bacterial endotoxin. Heat shock, but not other types of stress, significantly induced the transcription of this locus in ALL cells. Transient overexpression of cassini in human 293 embryonic kidney cells did not increase the cytotoxic or cytostatic effects of chemotherapeutic drugs but provided some protection. Database searches revealed that sequences highly homologous to cassini are present in rodents, apicomplexans, flatworms and primates, indicating that they are conserved in evolution. Moreover, CASSINI RNA was induced in human ALL cells treated with vincristine. Surprisingly, cassini belongs to the previously reported murine family of γ-satellite/major satellite DNA sequences, which were not known to be present in other species. CONCLUSIONS: Our results show that the transcription of at least one member of these sequences is regulated, suggesting that this has a function in normal and transformed immune cells. Expression of these sequences may protect cells when they are exposed to specific stress stimuli.


Subject(s)
Proteins/metabolism , RNA/metabolism , Animals , Antineoplastic Agents, Phytogenic/toxicity , Apicomplexa , Biological Evolution , Cell Line, Tumor , Coculture Techniques , Databases, Genetic , Endotoxins/toxicity , Fibroblasts/metabolism , Gene Expression Profiling , Genetic Loci , HEK293 Cells , Humans , Mice , Mice, Transgenic , Platyhelminths , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Primates , Proteins/genetics , Pyrimidines/toxicity , RNA/analysis , RNA/genetics , Temperature , Transfection , Up-Regulation/drug effects , Vincristine/toxicity
16.
J Exp Med ; 201(11): 1837-52, 2005 Jun 06.
Article in English | MEDLINE | ID: mdl-15939795

ABSTRACT

Pre-B cells undergo apoptosis unless they are rescued by pre-B cell receptor-dependent survival signals. We previously showed that the BCR-ABL1 kinase that is expressed in pre-B lymphoblastic leukemia bypasses selection for pre-B cell receptor-dependent survival signals. Investigating possible interference of BCR-ABL1 with pre-B cell receptor signaling, we found that neither SYK nor SLP65 can be phosphorylated in response to pre-B cell receptor engagement. Instead, Bruton's tyrosine kinase (BTK) is constitutively phosphorylated by BCR-ABL1. Activated BTK is essential for survival signals that otherwise would arise from the pre-B cell receptor, including activation of PLCgamma1, autonomous Ca2+ signaling, STAT5-phosphorylation, and up-regulation of BCLX(L). Inhibition of BTK activity specifically induces apoptosis in BCR-ABL1+ leukemia cells to a similar extent as inhibition of BCR-ABL1 kinase activity itself. However, BCR-ABL1 cannot directly bind to full-length BTK. Instead, BCR-ABL1 induces the expression of a truncated splice variant of BTK that acts as a linker between the two kinases. As opposed to full-length BTK, truncated BTK lacks kinase activity yet can bind to BCR-ABL1 through its SRC-homology domain 3. Acting as a linker, truncated BTK enables BCR-ABL1-dependent activation of full-length BTK, which initiates downstream survival signals and mimics a constitutively active pre-B cell receptor.


Subject(s)
Calcium Signaling , Gene Expression Regulation, Leukemic , Membrane Glycoproteins/metabolism , Molecular Mimicry , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adolescent , Adult , Aged , Cell Line, Tumor , Cell Survival , Child , Child, Preschool , Female , Humans , Male , Membrane Glycoproteins/immunology , Middle Aged , Pre-B Cell Receptors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell
17.
Nat Commun ; 12(1): 512, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479235

ABSTRACT

To achieve replicative immortality, cancer cells must activate telomere maintenance mechanisms to prevent telomere shortening. ~85% of cancers circumvent telomeric attrition by re-expressing telomerase, while the remaining ~15% of cancers induce alternative lengthening of telomeres (ALT), which relies on break-induced replication (BIR) and telomere recombination. Although ALT tumours were first reported over 20 years ago, the mechanism of ALT induction remains unclear and no study to date has described a cell-based model that permits the induction of ALT. Here, we demonstrate that infection with Kaposi's sarcoma herpesvirus (KSHV) induces sustained acquisition of ALT-like features in previously non-ALT cell lines. KSHV-infected cells acquire hallmarks of ALT activity that are also observed in KSHV-associated tumour biopsies. Down-regulating BIR impairs KSHV latency, suggesting that KSHV co-opts ALT for viral functionality. This study uncovers KSHV infection as a means to study telomere maintenance by ALT and reveals features of ALT in KSHV-associated tumours.


Subject(s)
Neoplasms/genetics , Telomere Homeostasis/genetics , Telomere Shortening/genetics , Telomere/genetics , Carcinogenesis , Cell Line , Cell Line, Tumor , DNA Damage , DNA Replication/genetics , HeLa Cells , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Humans , In Situ Hybridization, Fluorescence , Neoplasms/pathology , Neoplasms/virology , Proteome/genetics , Proteome/metabolism , Telomerase/genetics , Telomerase/metabolism
18.
J Exp Med ; 199(5): 673-85, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14993251

ABSTRACT

The BCR-ABL1 kinase expressed in acute lymphoblastic leukemia (ALL) drives malignant transformation of human pre-B cells. Comparing genome-wide gene expression profiles of BCR-ABL1+ pre-B ALL and normal bone marrow pre-B cells by serial analysis of gene expression, many genes involved in pre-B cell receptor signaling are silenced in the leukemia cells. Although normal pre-B cells are selected for the expression of a functional pre-B cell receptor, BCR-ABL1+ ALL cells mostly do not harbor a productively rearranged IGH allele. In these cases, we identified traces of secondary VH gene rearrangements, which may have rendered an initially productive VH region gene nonfunctional. Even BCR-ABL1+ ALL cells harboring a functional VH region gene are unresponsive to pre-B cell receptor engagement and exhibit autonomous oscillatory Ca2+ signaling activity. Conversely, leukemia subclones surviving inhibition of BCR-ABL1 by STI571 restore responsiveness to antigen receptor engagement and differentiate into immature B cells expressing immunoglobulin light chains. BCR-ABL1 kinase activity is linked to defective pre-B cell receptor signaling and the expression of a truncated isoform of the pre-B cell receptor-associated linker molecule SLP65. Also in primary leukemia cells, truncated SLP65 is expressed before but not after treatment of the patients with STI571. We conclude that inhibition of BCR-ABL1 reconstitutes selection for leukemia cells expressing a functional (pre-) B cell receptor.


Subject(s)
Membrane Glycoproteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Protein-Tyrosine Kinases/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Base Sequence , Carrier Proteins/genetics , Child , Child, Preschool , DNA, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl , Gene Expression , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , Male , Middle Aged , Phosphoproteins/genetics , Pre-B Cell Receptors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Receptors, Antigen, B-Cell , Selection, Genetic
19.
J Exp Med ; 196(10): 1291-305, 2002 Nov 18.
Article in English | MEDLINE | ID: mdl-12438421

ABSTRACT

To identify changes in the regulation of B cell receptor (BCR) signals during the development of human B cells, we generated genome-wide gene expression profiles using the serial analysis of gene expression (SAGE) technique for CD34(+) hematopoietic stem cells (HSCs), pre-B cells, naive, germinal center (GC), and memory B cells. Comparing these SAGE profiles, genes encoding positive regulators of BCR signaling were expressed at consistently lower levels in naive B cells than in all other B cell subsets. Conversely, a large group of inhibitory signaling molecules, mostly belonging to the immunoglobulin superfamily (IgSF), were specifically or predominantly expressed in naive B cells. The quantitative differences observed by SAGE were corroborated by semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry. In a functional assay, we show that down-regulation of inhibitory IgSF receptors and increased responsiveness to BCR stimulation in memory as compared with naive B cells at least partly results from interleukin (IL)-4 receptor signaling. Conversely, activation or impairment of the inhibitory IgSF receptor LIRB1 affected BCR-dependent Ca(2+) mobilization only in naive but not memory B cells. Thus, LIRB1 and IL-4 may represent components of two nonoverlapping gene expression programs in naive and memory B cells, respectively: in naive B cells, a large group of inhibitory IgSF receptors can elevate the BCR signaling threshold to prevent these cells from premature activation and clonal expansion before GC-dependent affinity maturation. In memory B cells, facilitated responsiveness upon reencounter of the immunizing antigen may result from amplification of BCR signals at virtually all levels of signal transduction.


Subject(s)
B-Lymphocytes/immunology , Receptors, Antigen, B-Cell/immunology , Antigens, CD34/immunology , Gene Expression Profiling , Humans , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
20.
J Exp Med ; 200(2): 123-35, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15263023

ABSTRACT

Here a new, intrinsically pluripotent, CD45-negative population from human cord blood, termed unrestricted somatic stem cells (USSCs) is described. This rare population grows adherently and can be expanded to 10(15) cells without losing pluripotency. In vitro USSCs showed homogeneous differentiation into osteoblasts, chondroblasts, adipocytes, and hematopoietic and neural cells including astrocytes and neurons that express neurofilament, sodium channel protein, and various neurotransmitter phenotypes. Stereotactic implantation of USSCs into intact adult rat brain revealed that human Tau-positive cells persisted for up to 3 mo and showed migratory activity and a typical neuron-like morphology. In vivo differentiation of USSCs along mesodermal and endodermal pathways was demonstrated in animal models. Bony reconstitution was observed after transplantation of USSC-loaded calcium phosphate cylinders in nude rat femurs. Chondrogenesis occurred after transplanting cell-loaded gelfoam sponges into nude mice. Transplantation of USSCs in a noninjury model, the preimmune fetal sheep, resulted in up to 5% human hematopoietic engraftment. More than 20% albumin-producing human parenchymal hepatic cells with absence of cell fusion and substantial numbers of human cardiomyocytes in both atria and ventricles of the sheep heart were detected many months after USSC transplantation. No tumor formation was observed in any of these animals.


Subject(s)
Cell Line , Fetal Blood/cytology , Placenta/blood supply , Stem Cells/cytology , Adipocytes/cytology , Albumins/metabolism , Animals , Blotting, Western , Bone and Bones/cytology , Cell Culture Techniques , Cell Differentiation , Cell Division , Cell Transplantation , Cord Blood Stem Cell Transplantation , Femur/metabolism , Flow Cytometry , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Hippocampus/cytology , Humans , Immunophenotyping , Leukocyte Common Antigens/biosynthesis , Leukocytes, Mononuclear/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Neurotransmitter Agents , Osteoblasts/metabolism , Phenotype , Polymerase Chain Reaction , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Time Factors , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL