Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Trends Immunol ; 42(5): 371-374, 2021 05.
Article in English | MEDLINE | ID: mdl-33773925

ABSTRACT

The prevalence of childhood B cell acute lymphoblastic leukemia (B-ALL) is increasing, particularly in developed countries. There is no clear explanation for this increment, but recent data suggest that, besides genetic predisposition, stress in the immune system (e.g., an infection) might have an important role in B-ALL leukemogenesis. Here, we speculate on how this knowledge might impact B-ALL prevention strategies.


Subject(s)
Leukemia, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Genetic Predisposition to Disease , Humans
2.
Blood ; 137(13): 1741-1753, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33024996

ABSTRACT

Diffuse large B-cell lymphomas (DLBCLs) are clinically and genetically heterogeneous tumors. Deregulation of diverse biological processes specific to B cells, such as B-cell receptor (BCR) signaling and motility regulation, contribute to lymphomagenesis. Human germinal center associated lymphoma (HGAL) is a B-cell-specific adaptor protein controlling BCR signaling and B lymphocyte motility. In normal B cells, it is expressed in germinal center (GC) B lymphocytes and promptly downregulated upon further differentiation. The majority of DLBCL tumors, primarily GC B-cell types, but also activated types, express HGAL. To investigate the consequences of constitutive expression of HGAL in vivo, we generated mice that conditionally express human HGAL at different stages of hematopoietic development using 3 restricted Cre-mediated approaches to initiate expression of HGAL in hematopoietic stem cells, pro-B cells, or GC B cells. Following immune stimulation, we observed larger GCs in mice in which HGAL expression was initiated in GC B cells. All 3 mouse strains developed DLBCL at a frequency of 12% to 30% starting at age 13 months, leading to shorter survival. Immunohistochemical studies showed that all analyzed tumors were of the GC B-cell type. Exon sequencing revealed mutations reported in human DLBCL. Our data demonstrate that constitutive enforced expression of HGAL leads to DLBCL development.


Subject(s)
Carcinogenesis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Microfilament Proteins/genetics , Animals , Carcinogenesis/pathology , Cell Line , Female , Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Germinal Center/metabolism , Germinal Center/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred C57BL
3.
Int J Mol Sci ; 24(14)2023 Jul 16.
Article in English | MEDLINE | ID: mdl-37511301

ABSTRACT

Cancer stem cells (CSCs) are now well-established as key players in tumor initiation, progression, and therapy resistance [...].


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology
4.
EMBO J ; 37(14)2018 07 13.
Article in English | MEDLINE | ID: mdl-29880602

ABSTRACT

The impact of LMO2 expression on cell lineage decisions during T-cell leukemogenesis remains largely elusive. Using genetic lineage tracing, we have explored the potential of LMO2 in dictating a T-cell malignant phenotype. We first initiated LMO2 expression in hematopoietic stem/progenitor cells and maintained its expression in all hematopoietic cells. These mice develop exclusively aggressive human-like T-ALL In order to uncover a potential exclusive reprogramming effect of LMO2 in murine hematopoietic stem/progenitor cells, we next showed that transient LMO2 expression is sufficient for oncogenic function and induction of T-ALL The resulting T-ALLs lacked LMO2 and its target-gene expression, and histologically, transcriptionally, and genetically similar to human LMO2-driven T-ALL We next found that during T-ALL development, secondary genomic alterations take place within the thymus. However, the permissiveness for development of T-ALL seems to be associated with wider windows of differentiation than previously appreciated. Restricted Cre-mediated activation of Lmo2 at different stages of B-cell development induces systematically and unexpectedly T-ALL that closely resembled those of their natural counterparts. Together, these results provide a novel paradigm for the generation of tumor T cells through reprogramming in vivo and could be relevant to improve the response of T-ALL to current therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis , Cell Transformation, Neoplastic , LIM Domain Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Disease Models, Animal , Gene Expression Profiling , Hematopoietic Stem Cells/physiology , Histocytochemistry , Mice , Thymus Gland/pathology
5.
Blood ; 136(18): 2003-2017, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32911536

ABSTRACT

The majority of childhood leukemias are precursor B-cell acute lymphoblastic leukemias (pB-ALLs) caused by a combination of prenatal genetic predispositions and oncogenic events occurring after birth. Although genetic predispositions are frequent in children (>1% to 5%), fewer than 1% of genetically predisposed carriers will develop pB-ALL. Although infectious stimuli are believed to play a major role in leukemogenesis, the critical determinants are not well defined. Here, by using murine models of pB-ALL, we show that microbiome disturbances incurred by antibiotic treatment early in life were sufficient to induce leukemia in genetically predisposed mice, even in the absence of infectious stimuli and independent of T cells. By using V4 and full-length 16S ribosomal RNA sequencing of a series of fecal samples, we found that genetic predisposition to pB-ALL (Pax5 heterozygosity or ETV6-RUNX1 fusion) shaped a distinct gut microbiome. Machine learning accurately (96.8%) predicted genetic predisposition using 40 of 3983 amplicon sequence variants as proxies for bacterial species. Transplantation of either wild-type (WT) or Pax5+/- hematopoietic bone marrow cells into WT recipient mice revealed that the microbiome is shaped and determined in a donor genotype-specific manner. Gas chromatography-mass spectrometry (GC-MS) analyses of sera from WT and Pax5+/- mice demonstrated the presence of a genotype-specific distinct metabolomic profile. Taken together, our data indicate that it is a lack of commensal microbiota rather than the presence of specific bacteria that promotes leukemia in genetically predisposed mice. Future large-scale longitudinal studies are required to determine whether targeted microbiome modification in children predisposed to pB-ALL could become a successful prevention strategy.


Subject(s)
Disease Susceptibility , Dysbiosis/complications , Feces/microbiology , Gastrointestinal Microbiome , Leukemia, Experimental/prevention & control , PAX5 Transcription Factor/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Animals , Female , Leukemia, Experimental/genetics , Leukemia, Experimental/microbiology , Leukemia, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/microbiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
6.
Int J Mol Sci ; 23(14)2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35886910

ABSTRACT

Leukemia is the most usual childhood cancer, and B-cell acute lymphoblastic leukemia (B-ALL) is its most common presentation. It has been proposed that pediatric leukemogenesis occurs through a "multi-step" or "multi-hit" mechanism that includes both in utero and postnatal steps. Many childhood leukemia-initiating events, such as chromosomal translocations, originate in utero, and studies so far suggest that these "first-hits" occur at a far higher frequency than the incidence of childhood leukemia itself. The reason why only a small percentage of the children born with such preleukemic "hits" will develop full-blown leukemia is still a mystery. In order to better understand childhood leukemia, mouse modeling is essential, but only if the multistage process of leukemia can be recapitulated in the model. Therefore, mouse models naturally reproducing the "multi-step" process of childhood B-ALL will be essential to identify environmental or other factors that are directly linked to increased risk of disease.


Subject(s)
Myelodysplastic Syndromes , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Preleukemia , Animals , Disease Models, Animal , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Preleukemia/genetics , Translocation, Genetic
7.
Blood ; 129(19): 2645-2656, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28288979

ABSTRACT

CREBBP is targeted by inactivating mutations in follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL). Here, we provide evidence from transgenic mouse models that Crebbp deletion results in deficits in B-cell development and can cooperate with Bcl2 overexpression to promote B-cell lymphoma. Through transcriptional and epigenetic profiling of these B cells, we found that Crebbp inactivation was associated with broad transcriptional alterations, but no changes in the patterns of histone acetylation at the proximal regulatory regions of these genes. However, B cells with Crebbp inactivation showed high expression of Myc and patterns of altered histone acetylation that were localized to intragenic regions, enriched for Myc DNA binding motifs, and showed Myc binding. Through the analysis of CREBBP mutations from a large cohort of primary human FL and DLBCL, we show a significant difference in the spectrum of CREBBP mutations in these 2 diseases, with higher frequencies of nonsense/frameshift mutations in DLBCL compared with FL. Together, our data therefore provide important links between Crebbp inactivation and Bcl2 dependence and show a role for Crebbp inactivation in the induction of Myc expression. We suggest this may parallel the role of CREBBP frameshift/nonsense mutations in DLBCL that result in loss of the protein, but may contrast the role of missense mutations in the lysine acetyltransferase domain that are more frequently observed in FL and yield an inactive protein.


Subject(s)
B-Lymphocytes/pathology , CREB-Binding Protein/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Epigenesis, Genetic , Gene Deletion , Humans , Lymphoma, Follicular/genetics , Mice , Mice, Transgenic , Mutation
8.
Bioelectromagnetics ; 40(5): 343-353, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31157932

ABSTRACT

Exposure to extremely low-frequency magnetic fields (ELF-MFs) has been classified by the International Agency for Research on Cancer (IARC) as "possibly carcinogenic to humans," based on limited scientific evidence concerning childhood leukemia. This assessment emphasized the lack of appropriate animal models recapitulating the natural history of this disease. Childhood B-cell acute lymphoblastic leukemia (B-ALL) is the result of complex interactions between genetic susceptibility and exposure to exogenous agents. The most common chromosomal alteration is the ETV6-RUNX1 fusion gene, which confers a low risk of developing the malignancy by originating a preleukemic clone requiring secondary hits for full-blown disease to appear. To develop potential prophylactic interventions, we need to identify the environmental triggers of the second hit. Recently, we generated a B-ALL mouse model of the human ETV6-RUNX1+ preleukemic state. Here, we present the results from the ARIMMORA pilot study, obtained by exposing 34 Sca1-ETV6-RUNX1 mice (vs. 27 unexposed) to a 50 Hz magnetic field of 1.5 mT with both fundamental and harmonic content, with an on/off cycle of 10 min/5 min, for 20 h/day, from conception until 3 months of age. Mice were monitored until 2 years of age and peripheral blood was periodically analyzed by flow cytometry. One of the exposed mice developed B-ALL while none of the non-exposed did. Although the results are statistically non-significant due to the limited number of mice used in this pilot experiment, overall, the results show that the newly developed Sca1-ETV6-RUNX1 mouse can be successfully used for ELF-MF exposure studies about the etiology of childhood B-ALL. Bioelectromagnetics. 2019;40:343-353. © 2019 Bioelectromagnetics Society.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Disease Models, Animal , Electromagnetic Fields/adverse effects , Leukemia, Experimental , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Proteins c-ets/genetics , Radio Waves/adverse effects , Repressor Proteins/genetics , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Pilot Projects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , ETS Translocation Variant 6 Protein
9.
Proc Natl Acad Sci U S A ; 112(10): E1116-25, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713363

ABSTRACT

Follicular lymphoma (FL) is incurable with conventional therapies and has a clinical course typified by multiple relapses after therapy. These tumors are genetically characterized by B-cell leukemia/lymphoma 2 (BCL2) translocation and mutation of genes involved in chromatin modification. By analyzing purified tumor cells, we identified additional novel recurrently mutated genes and confirmed mutations of one or more chromatin modifier genes within 96% of FL tumors and two or more in 76% of tumors. We defined the hierarchy of somatic mutations arising during tumor evolution by analyzing the phylogenetic relationship of somatic mutations across the coding genomes of 59 sequentially acquired biopsies from 22 patients. Among all somatically mutated genes, CREBBP mutations were most significantly enriched within the earliest inferable progenitor. These mutations were associated with a signature of decreased antigen presentation characterized by reduced transcript and protein abundance of MHC class II on tumor B cells, in line with the role of CREBBP in promoting class II transactivator (CIITA)-dependent transcriptional activation of these genes. CREBBP mutant B cells stimulated less proliferation of T cells in vitro compared with wild-type B cells from the same tumor. Transcriptional signatures of tumor-infiltrating T cells were indicative of reduced proliferation, and this corresponded to decreased frequencies of tumor-infiltrating CD4 helper T cells and CD8 memory cytotoxic T cells. These observations therefore implicate CREBBP mutation as an early event in FL evolution that contributes to immune evasion via decreased antigen presentation.


Subject(s)
Antigen-Presenting Cells/immunology , Lymphoma, Follicular/genetics , Mutation , Neoplastic Stem Cells/pathology , CREB-Binding Protein/genetics , Chromatin/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Humans , Lymphoma, Follicular/immunology , Polymerase Chain Reaction
10.
Int J Mol Sci ; 19(5)2018 May 17.
Article in English | MEDLINE | ID: mdl-29772764

ABSTRACT

Due to the clonal nature of human leukemia evolution, all leukemic cells carry the same leukemia-initiating genetic lesions, independently of the intrinsic tumoral cellular heterogeneity. However, the latest findings have shown that the mode of action of oncogenes is not homogeneous throughout the developmental history of leukemia. Studies on different types of hematopoietic tumors have shown that the contribution of oncogenes to leukemia is mainly mediated through the epigenetic reprogramming of the leukemia-initiating target cell. This driving of cancer by a malignant epigenetic stem cell rewiring is, however, not exclusive of the hematopoietic system, but rather represents a common tumoral mechanism that is also at work in epithelial tumors. Tumoral epigenetic reprogramming is therefore a new type of interaction between genes and their target cells, in which the action of the oncogene modifies the epigenome to prime leukemia development by establishing a new pathological tumoral cellular identity. This reprogramming may remain latent until it is triggered by either endogenous or environmental stimuli. This new view on the making of leukemia not only reveals a novel function for oncogenes, but also provides evidence for a previously unconsidered model of leukemogenesis, in which the programming of the leukemia cellular identity has already occurred at the level of stem cells, therefore showing a role for oncogenes in the timing of leukemia initiation.


Subject(s)
Leukemia/etiology , Leukemia/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming , Environment , Epigenesis, Genetic , Genetic Predisposition to Disease , Hematopoiesis/genetics , Humans , Leukemia/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogenes
11.
EMBO J ; 32(11): 1502-13, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23632857

ABSTRACT

Tumour-associated oncogenes induce unscheduled proliferation as well as genomic and chromosomal instability. According to current models, therapeutic strategies that block oncogene activity are likely to selectively target tumour cells. However, recent evidences have revealed that oncogenes are only essential for the proliferation of some specific tumour cell types, but not all. Indeed, the latest studies of the interactions between the oncogene and its target cell have shown that oncogenes contribute to cancer development not only by inducing proliferation but also by developmental reprogramming of the epigenome. This provides the first evidence that tumorigenesis can be initiated by stem cell reprogramming, and uncovers a new role for oncogenes in the origin of cancer. Here we analyse these evidences and propose an updated model of oncogene function that can explain the full range of genotype-phenotype associations found in human cancer. Finally, we discuss how this vision opens new avenues for developing novel anti-cancer interventions.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genetic Association Studies , Neoplasms/genetics , Oncogenes/physiology , Animals , Cell Biology , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Chromosomal Instability , Humans , Mice , Models, Biological , Mutation , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells , Oncogenes/genetics
12.
Proc Natl Acad Sci U S A ; 111(1): 255-60, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24367082

ABSTRACT

Ubiquitous expression of a resident K-Ras(G12V) oncogene in adult mice revealed that most tissues are resistant to K-Ras oncogenic signals. Indeed, K-Ras(G12V) expression only induced overt tumors in lungs. To identify these transformation-permissive cells, we induced K-Ras(G12V) expression in a very limited number of adult lung cells (0.2%) and monitored their fate by X-Gal staining, a surrogate marker coexpressed with the K-Ras(G12V) oncoprotein. Four weeks later, 30% of these cells had proliferated to form small clusters. However, only SPC(+) alveolar type II (ATII) cells were able to form hyperplastic lesions, some of which progressed to adenomas and adenocarcinomas. In contrast, induction of K-Ras(G12V) expression in lung cells by intratracheal infection with adenoviral-Cre particles generated hyperplasias in all regions except the proximal airways. Bronchiolar and bronchioalveolar duct junction hyperplasias were primarily made of CC10(+) Clara cells. Some of them progressed to form benign adenomas. However, only alveolar hyperplasias, exclusively made up of SPC(+) ATII cells, progressed to yield malignant adenocarcinomas. Adenoviral infection induced inflammatory infiltrates primarily made of T and B cells. This inflammatory response was essential for the development of K-Ras(G12V)-driven bronchiolar hyperplasias and adenomas, but not for the generation of SPC(+) ATII lesions. Finally, activation of K-Ras(G12V) during embryonic development under the control of a Sca1 promoter yielded CC10(+), but not SPC(+), hyperplasias, and adenomas. These results, taken together, illustrate that different types of lung cells can generate benign lesions in response to K-Ras oncogenic signals. However, in adult mice, only SPC(+) ATII cells were able to yield malignant adenocarcinomas.


Subject(s)
Adenocarcinoma/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Lung Neoplasms/metabolism , Lung/cytology , ras Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Adenoviridae/metabolism , Alleles , Animals , Bronchioles/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , Cell Separation , Cell Transformation, Neoplastic , Flow Cytometry , Gene Expression Profiling , Inflammation , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Oncogenes , Promoter Regions, Genetic , Pulmonary Alveoli/metabolism , Signal Transduction , Stem Cells/cytology
13.
Semin Cancer Biol ; 32: 3-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24530939

ABSTRACT

Cancer is a clonal malignant disease originated in a single cell and characterized by the accumulation of partially differentiated cells that are phenotypically reminiscent of normal stages of differentiation. According to current models, therapeutic strategies that block oncogene activity are likely to selectively target tumor cells. However, recent evidences have revealed that cancer stem cells could arise through a tumor stem cell reprogramming mechanism, suggesting that genetic lesions that initiate the cancer process might be dispensable for tumor progression and maintenance. This review addresses the impact of these results toward a better understanding of cancer development and proposes new approaches to treat cancer in the future.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Reprogramming/genetics , Neoplasms/genetics , Neoplastic Stem Cells , Oncogenes/genetics , Animals , Cell Differentiation/genetics , Disease Models, Animal , Humans , Mice , Models, Biological , Neoplasms/therapy
14.
EMBO J ; 31(18): 3704-17, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22903061

ABSTRACT

Understanding the cellular origin of cancer can help to improve disease prevention and therapeutics. Human plasma cell neoplasias are thought to develop from either differentiated B cells or plasma cells. However, when the expression of Maf oncogenes (associated to human plasma cell neoplasias) is targeted to mouse B cells, the resulting animals fail to reproduce the human disease. Here, to explore early cellular changes that might take place in the development of plasma cell neoplasias, we engineered transgenic mice to express MafB in haematopoietic stem/progenitor cells (HS/PCs). Unexpectedly, we show that plasma cell neoplasias arise in the MafB-transgenic mice. Beyond their clinical resemblance to human disease, these neoplasias highly express genes that are known to be upregulated in human multiple myeloma. Moreover, gene expression profiling revealed that MafB-expressing HS/PCs were more similar to B cells and tumour plasma cells than to any other subset, including wild-type HS/PCs. Consistent with this, genome-scale DNA methylation profiling revealed that MafB imposes an epigenetic program in HS/PCs, and that this program is preserved in mature B cells of MafB-transgenic mice, demonstrating a novel molecular mechanism involved in tumour initiation. Our findings suggest that, mechanistically, the haematopoietic progenitor population can be the target for transformation in MafB-associated plasma cell neoplasias.


Subject(s)
Gene Expression Regulation, Neoplastic , MafB Transcription Factor/metabolism , Multiple Myeloma/metabolism , Animals , Antigens, CD34/biosynthesis , Antigens, Ly/metabolism , B-Lymphocytes/metabolism , DNA Methylation , DNA, Complementary/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Library , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization, Fluorescence , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Translocation, Genetic
15.
Int J Mol Sci ; 17(6)2016 Jun 06.
Article in English | MEDLINE | ID: mdl-27275819

ABSTRACT

Leukemic stem cells (LSCs) are defined as cells that possess the ability to self-renew and give rise to the differentiated cancer cells that comprise the tumor. These LSCs seem to show chemo-resistance and radio-resistance leading to the failure of conventional cancer therapies. Current therapies are directed at the fast growing tumor mass leaving the LSC fraction untouched. Eliminating LSCs, the root of cancer origin and recurrence, is considered to be a hopeful approach to improve survival or even to cure cancer patients. In order to achieve this, the characterization of LSCs is a prerequisite in order to develop LSC-based therapies to eliminate them. Here we review if vitamin D analogues may allow an avenue to target the LSCs.


Subject(s)
Leukemia/drug therapy , Leukemia/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Vitamin D/pharmacology , Vitamin D/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Epigenesis, Genetic/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/genetics , Molecular Targeted Therapy , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives
16.
Proc Natl Acad Sci U S A ; 109(26): 10534-9, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22689981

ABSTRACT

Chromosomal translocations involving the MALT1 gene are hallmarks of mucosa-associated lymphoid tissue (MALT) lymphoma. To date, targeting these translocations to mouse B cells has failed to reproduce human disease. Here, we induced MALT1 expression in mouse Sca1(+)Lin(-) hematopoietic stem/progenitor cells, which showed NF-κB activation and early lymphoid priming, being selectively skewed toward B-cell differentiation. These cells accumulated in extranodal tissues and gave rise to clonal tumors recapitulating the principal clinical, biological, and molecular genetic features of MALT lymphoma. Deletion of p53 gene accelerated tumor onset and induced transformation of MALT lymphoma to activated B-cell diffuse large-cell lymphoma (ABC-DLBCL). Treatment of MALT1-induced lymphomas with a specific inhibitor of MALT1 proteolytic activity decreased cell viability, indicating that endogenous Malt1 signaling was required for tumor cell survival. Our study shows that human-like lymphomas can be modeled in mice by targeting MALT1 expression to hematopoietic stem/progenitor cells, demonstrating the oncogenic role of MALT1 in lymphomagenesis. Furthermore, this work establishes a molecular link between MALT lymphoma and ABC-DLBCL, and provides mouse models to test MALT1 inhibitors. Finally, our results suggest that hematopoietic stem/progenitor cells may be involved in the pathogenesis of human mature B-cell lymphomas.


Subject(s)
Caspases/genetics , Hematopoietic Stem Cells/metabolism , Lymphoma/pathology , Neoplasm Proteins/genetics , Oncogenes , Animals , Humans , Mice , Mice, Transgenic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Transcription, Genetic
17.
Biol Chem ; 395(11): 1315-20, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205718

ABSTRACT

Abstract A cancer dogma states that inactivation of oncogene(s) can cause cancer remission, implying that oncogenes are the Achilles' heel of cancers. This current model of cancer has kept oncogenes firmly in focus as therapeutic targets and is in agreement with the fact that in human cancers all cancerous cells, with independence of the cellular heterogeneity existing within the tumour, carry the same oncogenic genetic lesions. However, recent studies of the interactions between an oncogene and its target cell have shown that oncogenes contribute to cancer development via developmental reprogramming of the epigenome within the target cell. These results provide the first evidence that carcinogenesis can be initiated by epigenetic stem cell reprogramming, and uncover a new role for oncogenes in the origin of cancer. Here we analyse these evidences and discuss how this vision offers new avenues for developing novel anti-cancer interventions.


Subject(s)
Epigenesis, Genetic , Neoplasms/genetics , Oncogenes , Animals , Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
18.
EMBO J ; 28(1): 8-20, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19037256

ABSTRACT

In human cancers, all cancerous cells carry the oncogenic genetic lesions. However, to elucidate whether cancer is a stem cell-driven tissue, we have developed a strategy to limit oncogene expression to the stem cell compartment in a transgenic mouse setting. Here, we focus on the effects of the BCR-ABLp210 oncogene, associated with chronic myeloid leukaemia (CML) in humans. We show that CML phenotype and biology can be established in mice by restricting BCR-ABLp210 expression to stem cell antigen 1 (Sca1)(+) cells. The course of the disease in Sca1-BCR-ABLp210 mice was not modified on STI571 treatment. However, BCR-ABLp210-induced CML is reversible through the unique elimination of the cancer stem cells (CSCs). Overall, our data show that oncogene expression in Sca1(+) cells is all that is required to fully reprogramme it, giving rise to a full-blown, oncogene-specified tumour with all its mature cellular diversity, and that elimination of the CSCs is enough to eradicate the whole tumour.


Subject(s)
Gene Expression , Genes, abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Stem Cells , Animals , Ataxin-1 , Ataxins , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/analysis , Nuclear Proteins/analysis , Survival Analysis
19.
Front Immunol ; 14: 1285743, 2023.
Article in English | MEDLINE | ID: mdl-37901253

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) stands as the primary contributor to childhood cancer-related mortality on a global scale. The development of the most conventional forms of this disease has been proposed to be conducted by two different steps influenced by different types of risk factors. The first step is led by a genetic insult that is presumably acquired before birth that transforms a healthy cell into a preleukemic one, which is maintained untransformed until the second step takes place. This necessary next step to leukemia development will be triggered by different risk factors to which children are exposed after birth. Murine models that recap the stepwise progression of B-ALL have been instrumental in identifying environmental and genetic factors that contribute to disease risk. Recent evidence from these models has demonstrated that specific environmental risk factors, such as common infections or gut microbiome dysbiosis, induce immune stress, driving the transformation of preleukemic cells, and harboring genetic alterations, into fully transformed leukemic cells. Such models serve as valuable tools for investigating the mechanisms underlying preleukemic events and can aid in the development of preventive approaches for leukemia in child. Here, we discuss the existing knowledge, learned from mouse models, of the impact of genetic and environmental risk factors on childhood B-ALL evolution and how B-ALL prevention could be reached by interfering with preleukemic cells.


Subject(s)
Leukemia, B-Cell , Leukemia, Lymphocytic, Chronic, B-Cell , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Mice , Animals , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Risk Factors
20.
Nat Commun ; 14(1): 5159, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620322

ABSTRACT

The initial steps of B-cell acute lymphoblastic leukemia (B-ALL) development usually pass unnoticed in children. Several preclinical studies have shown that exposure to immune stressors triggers the transformation of preleukemic B cells to full-blown B-ALL, but how this takes place is still a longstanding and unsolved challenge. Here we show that dysregulation of innate immunity plays a driving role in the clonal evolution of pre-malignant Pax5+/- B-cell precursors toward leukemia. Transcriptional profiling reveals that Myd88 is downregulated in immune-stressed pre-malignant B-cell precursors and in leukemic cells. Genetic reduction of Myd88 expression leads to a significant increase in leukemia incidence in Pax5+/-Myd88+/- mice through an inflammation-dependent mechanism. Early induction of Myd88-independent Toll-like receptor 3 signaling results in a significant delay of leukemia development in Pax5+/- mice. Altogether, these findings identify a role for innate immunity dysregulation in leukemia, with important implications for understanding and therapeutic targeting of the preleukemic state in children.


Subject(s)
Burkitt Lymphoma , Leukemia , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Precursor Cells, B-Lymphoid , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing , Immunity, Innate , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL