Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 16(7): 766-774, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25985233

ABSTRACT

Childhood acute lymphoblastic leukemia (ALL) can often be traced to a pre-leukemic clone carrying a prenatal genetic lesion. Postnatally acquired mutations then drive clonal evolution toward overt leukemia. The enzymes RAG1-RAG2 and AID, which diversify immunoglobulin-encoding genes, are strictly segregated in developing cells during B lymphopoiesis and peripheral mature B cells, respectively. Here we identified small pre-BII cells as a natural subset with increased genetic vulnerability owing to concurrent activation of these enzymes. Consistent with epidemiological findings on childhood ALL etiology, susceptibility to genetic lesions during B lymphopoiesis at the transition from the large pre-BII cell stage to the small pre-BII cell stage was exacerbated by abnormal cytokine signaling and repetitive inflammatory stimuli. We demonstrated that AID and RAG1-RAG2 drove leukemic clonal evolution with repeated exposure to inflammatory stimuli, paralleling chronic infections in childhood.


Subject(s)
B-Lymphocytes/immunology , Clonal Evolution/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cells, B-Lymphoid/immunology , Adolescent , Animals , Antibody Diversity/genetics , Antibody Diversity/immunology , B-Lymphocytes/metabolism , Child , Child, Preschool , Clonal Evolution/genetics , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Female , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Infant , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Microscopy, Fluorescence , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
2.
N Engl J Med ; 374(23): 2209-2221, 2016 Jun 09.
Article in English | MEDLINE | ID: mdl-27276561

ABSTRACT

BACKGROUND: Recent studies have provided a detailed census of genes that are mutated in acute myeloid leukemia (AML). Our next challenge is to understand how this genetic diversity defines the pathophysiology of AML and informs clinical practice. METHODS: We enrolled a total of 1540 patients in three prospective trials of intensive therapy. Combining driver mutations in 111 cancer genes with cytogenetic and clinical data, we defined AML genomic subgroups and their relevance to clinical outcomes. RESULTS: We identified 5234 driver mutations across 76 genes or genomic regions, with 2 or more drivers identified in 86% of the patients. Patterns of co-mutation compartmentalized the cohort into 11 classes, each with distinct diagnostic features and clinical outcomes. In addition to currently defined AML subgroups, three heterogeneous genomic categories emerged: AML with mutations in genes encoding chromatin, RNA-splicing regulators, or both (in 18% of patients); AML with TP53 mutations, chromosomal aneuploidies, or both (in 13%); and, provisionally, AML with IDH2(R172) mutations (in 1%). Patients with chromatin-spliceosome and TP53-aneuploidy AML had poor outcomes, with the various class-defining mutations contributing independently and additively to the outcome. In addition to class-defining lesions, other co-occurring driver mutations also had a substantial effect on overall survival. The prognostic effects of individual mutations were often significantly altered by the presence or absence of other driver mutations. Such gene-gene interactions were especially pronounced for NPM1-mutated AML, in which patterns of co-mutation identified groups with a favorable or adverse prognosis. These predictions require validation in prospective clinical trials. CONCLUSIONS: The driver landscape in AML reveals distinct molecular subgroups that reflect discrete paths in the evolution of AML, informing disease classification and prognostic stratification. (Funded by the Wellcome Trust and others; ClinicalTrials.gov number, NCT00146120.).


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Adult , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA Mutational Analysis , Epistasis, Genetic , Gene Fusion , Genotype , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Middle Aged , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Proportional Hazards Models , Prospective Studies , RNA Splicing , Survival Analysis
3.
Haematologica ; 104(7): 1440-1450, 2019 07.
Article in English | MEDLINE | ID: mdl-30733268

ABSTRACT

The emergence of treatment resistant sub-clones is a key feature of relapse in multiple myeloma. Therapeutic attempts to extend remission and prevent relapse include maximizing response and the use of maintenance therapy. We used whole exome sequencing to study the genetics of paired samples taken at presentation and at relapse from 56 newly diagnosed patients, following induction therapy, randomized to receive either lenalidomide maintenance or observation as part of the Myeloma XI trial. Patients included were considered high risk, relapsing within 30 months of maintenance randomization. Patients achieving a complete response had predominantly branching evolutionary patterns leading to relapse, characterized by a greater mutational burden, an altered mutational profile, bi-allelic inactivation of tumor suppressor genes, and acquired structural aberrations. Conversely, in patients achieving a partial response, the evolutionary features were predominantly stable with a similar mutational and structural profile seen at both time points. There were no significant differences between patients relapsing after lenalidomide maintenance versus observation. This study shows that the depth of response is a key determinant of the evolutionary patterns seen at relapse. This trial is registered at clinicaltrials.gov identifier: 01554852.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Clonal Evolution , Multiple Myeloma/pathology , Mutation , Neoplasm Recurrence, Local/pathology , Aged , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lenalidomide/administration & dosage , Maintenance Chemotherapy , Male , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Remission Induction , Thalidomide/administration & dosage , Treatment Outcome , Exome Sequencing
4.
Nature ; 500(7463): 415-21, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23945592

ABSTRACT

All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, 'kataegis', is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutagenesis/genetics , Mutation/genetics , Neoplasms/genetics , Aging/genetics , Algorithms , Cell Transformation, Neoplastic/pathology , Cytidine Deaminase/genetics , DNA/genetics , DNA/metabolism , DNA Mutational Analysis , Humans , Models, Genetic , Mutagenesis, Insertional/genetics , Mutagens/pharmacology , Neoplasms/enzymology , Neoplasms/pathology , Organ Specificity , Reproducibility of Results , Sequence Deletion/genetics , Transcription, Genetic/genetics
5.
BMC Biol ; 16(1): 22, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29466995

ABSTRACT

Paraphrasing Dobzhansky's famous dictum, I discuss how interrogating cancer through the lens of evolution has transformed our understanding of its development, causality and treatment resistance. The emerging picture of cancer captures its extensive diversity and therapeutic resilience, highlighting the need for more innovative approaches to control.


Subject(s)
Biological Evolution , Mutation/physiology , Neoplasms/genetics , Neoplasms/metabolism , Selection, Genetic/physiology , Animals , Drug Resistance/physiology , Humans , Neoplasms/therapy
6.
Nature ; 481(7381): 306-13, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22258609

ABSTRACT

Cancers evolve by a reiterative process of clonal expansion, genetic diversification and clonal selection within the adaptive landscapes of tissue ecosystems. The dynamics are complex, with highly variable patterns of genetic diversity and resulting clonal architecture. Therapeutic intervention may destroy cancer clones and erode their habitats, but it can also inadvertently provide a potent selective pressure for the expansion of resistant variants. The inherently Darwinian character of cancer is the primary reason for this therapeutic failure, but it may also hold the key to more effective control.


Subject(s)
Clonal Evolution , Neoplasms/genetics , Neoplasms/pathology , Animals , Clone Cells/metabolism , Clone Cells/pathology , Genomics , Humans , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment
7.
Nature ; 469(7330): 356-61, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21160474

ABSTRACT

Little is known of the genetic architecture of cancer at the subclonal and single-cell level or in the cells responsible for cancer clone maintenance and propagation. Here we have examined this issue in childhood acute lymphoblastic leukaemia in which the ETV6-RUNX1 gene fusion is an early or initiating genetic lesion followed by a modest number of recurrent or 'driver' copy number alterations. By multiplexing fluorescence in situ hybridization probes for these mutations, up to eight genetic abnormalities can be detected in single cells, a genetic signature of subclones identified and a composite picture of subclonal architecture and putative ancestral trees assembled. Subclones in acute lymphoblastic leukaemia have variegated genetics and complex, nonlinear or branching evolutionary histories. Copy number alterations are independently and reiteratively acquired in subclones of individual patients, and in no preferential order. Clonal architecture is dynamic and is subject to change in the lead-up to a diagnosis and in relapse. Leukaemia propagating cells, assayed by serial transplantation in NOD/SCID IL2Rγ(null) mice, are also genetically variegated, mirroring subclonal patterns, and vary in competitive regenerative capacity in vivo. These data have implications for cancer genomics and for the targeted therapy of cancer.


Subject(s)
Clone Cells/pathology , Genetic Variation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Clone Cells/metabolism , Core Binding Factor Alpha 2 Subunit , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Disease Progression , Genotype , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oncogene Proteins, Fusion/genetics
8.
Adv Exp Med Biol ; 962: 217-228, 2017.
Article in English | MEDLINE | ID: mdl-28299660

ABSTRACT

Acute leukaemia is the major subtype of paediatric cancer with a cumulative risk of 1 in 2000 for children up to the age of 15 years. Childhood acute lymphoblastic leukaemia (ALL) is a biologically and clinically diverse disease with distinctive subtypes; multiple chromosomal translocations exist within the subtypes and each carries its own prognostic relevance. The most common chromosome translocation observed is the t(12;21) that results in an in-frame fusion between the first five exons of ETV6 (TEL) and almost the entire coding region of RUNX1 (AML1).The natural history of childhood ALL is almost entirely clinically silent and is well advanced at the point of diagnosis. It has, however, been possible to backtrack this process through molecular analysis of appropriate clinical samples: (i) leukaemic clones in monozygotic twins that are either concordant or discordant for ALL; (ii) archived neonatal blood spots or Guthrie cards from individuals who later developed leukaemia; and (iii) stored, viable cord blood cells.Here, we outline our studies on the aetiology and pathology of childhood ALL that provide molecular evidence for a monoclonal, prenatal origin of ETV6-RUNX1+ leukaemia in monozygotic identical twins. We provide mechanistic support for the concept that altered patterns of infection during early childhood can deliver the necessary promotional drive for the progression of ETV6-RUNX1+ pre-leukaemic cells into a postnatal overt leukaemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Humans , Oncogene Proteins, Fusion/genetics , Twins, Monozygotic , ETS Translocation Variant 6 Protein
9.
Genome Res ; 23(12): 2115-25, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056532

ABSTRACT

The development of cancer is a dynamic evolutionary process in which intraclonal, genetic diversity provides a substrate for clonal selection and a source of therapeutic escape. The complexity and topography of intraclonal genetic architectures have major implications for biopsy-based prognosis and for targeted therapy. High-depth, next-generation sequencing (NGS) efficiently captures the mutational load of individual tumors or biopsies. But, being a snapshot portrait of total DNA, it disguises the fundamental features of subclonal variegation of genetic lesions and of clonal phylogeny. Single-cell genetic profiling provides a potential resolution to this problem, but methods developed to date all have limitations. We present a novel solution to this challenge using leukemic cells with known mutational spectra as a tractable model. DNA from flow-sorted single cells is screened using multiplex targeted Q-PCR within a microfluidic platform allowing unbiased single-cell selection, high-throughput, and comprehensive analysis for all main varieties of genetic abnormalities: chimeric gene fusions, copy number alterations, and single-nucleotide variants. We show, in this proof-of-principle study, that the method has a low error rate and can provide detailed subclonal genetic architectures and phylogenies.


Subject(s)
Clonal Evolution , Genomics/methods , Mutation , Neoplasms/genetics , Phylogeny , Single-Cell Analysis , Cell Line, Tumor , DNA Copy Number Variations , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Multiplex Polymerase Chain Reaction , Polymorphism, Single Nucleotide
10.
Nat Rev Cancer ; 7(3): 213-21, 2007 03.
Article in English | MEDLINE | ID: mdl-17301845

ABSTRACT

Epidemiological, genetic and molecular biological studies have collectively provided us with a rich source of data that underpins our current understanding of the aetiology and molecular pathogenesis of cancer. But this perspective focuses on proximate mechanisms, and does not provide an adequate explanation for the prevalence of tumours and cancer in animal species or what seems to be the striking vulnerability of Homo sapiens. The central precept of Darwinian medicine is that vulnerability to cancer, and other major diseases, arises at least in part as a consequence of the 'design' limitations, compromises and trade-offs that characterize evolutionary processes.


Subject(s)
Biological Evolution , Neoplasms/epidemiology , Neoplasms/etiology , Selection, Genetic , Adaptation, Biological , Animals , Humans , Species Specificity
11.
Proc Natl Acad Sci U S A ; 110(18): 7429-33, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23569245

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the major pediatric cancer. At diagnosis, the developmental timing of mutations contributing critically to clonal diversification and selection can be buried in the leukemia's covert natural history. Concordance of ALL in monozygotic, monochorionic twins is a consequence of intraplacental spread of an initiated preleukemic clone. Studying monozygotic twins with ALL provides a unique means of uncovering the timeline of mutations contributing to clonal evolution, pre- and postnatally. We sequenced the whole genomes of leukemic cells from two twin pairs with ALL to comprehensively characterize acquired somatic mutations in ALL, elucidating the developmental timing of all genetic lesions. Shared, prenatal, coding-region single-nucleotide variants were limited to the putative initiating lesions. All other nonsynonymous single-nucleotide variants were distinct between tumors and, therefore, secondary and postnatal. These changes occurred in a background of noncoding mutational changes that were almost entirely discordant in twin pairs and likely passenger mutations acquired during leukemic cell proliferation.


Subject(s)
DNA Mutational Analysis , Genome, Human/genetics , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Child, Preschool , Humans , Time Factors
12.
Br J Haematol ; 171(4): 574-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26205622

ABSTRACT

Infant T-cell acute lymphoblastic leukaemia (iT-ALL) is a very rare and poorly defined entity with a poor prognosis. We assembled a unique series of 13 infants with T-ALL, which allowed us to identify genotypic abnormalities and to investigate prenatal origins. Matched samples (diagnosis/remission) were analysed by single nucleotide polymorphism-array to identify genomic losses and gains. In three cases, we identified a recurrent somatic deletion on chromosome 3. These losses result in the complete deletion of MLF1 and have not previously been described in T-ALL. We observed two cases with an 11p13 deletion (LMO2-related), one of which also harboured a deletion of RB1. Another case presented a large 11q14·1-11q23·2 deletion that included ATM and only five patients (38%) showed deletions of CDKN2A/B. Four cases showed NOTCH1 mutations; in one case FBXW7 was the sole mutation and three cases showed alterations in PTEN. KMT2A rearrangements (KMT2A-r) were detected in three out of 13 cases. For three patients, mutations and copy number alterations (including deletion of PTEN) could be backtracked to birth using neonatal blood spot DNA, demonstrating an in utero origin. Overall, our data indicates that iT-ALL has a diverse but distinctive profile of genotypic abnormalities when compared to T-ALL in older children and adults.


Subject(s)
Genotype , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Age of Onset , Aneuploidy , Base Sequence , Cell Cycle Proteins , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 3/ultrastructure , DNA Methylation , DNA, Neoplasm/genetics , DNA-Binding Proteins , Female , Fetal Diseases/genetics , Gene Deletion , Gene Dosage , Genes, Neoplasm , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Molecular Sequence Data , Mutation , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/embryology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Promoter Regions, Genetic/genetics , Proteins/genetics , Sequence Deletion
13.
Blood ; 122(19): 3298-307, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-23996088

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the major pediatric cancer diagnosed in economically developed countries with B-cell precursor (BCP)-ALL, accounting for approximately 70% of ALL. Recent genome-wide association studies (GWAS) have provided the first unambiguous evidence for common inherited susceptibility to BCP-ALL, identifying susceptibility loci at 7p12.2, 9p21.3, 10q21.2, and 14q11.2. To identify additional BCP-ALL susceptibility loci, we conducted a GWAS and performed a meta-analysis with a published GWAS totaling 1658 cases and 4723 controls, with validation in 1449 cases and 1488 controls. Combined analysis identified novel loci mapping to 10p12.2 (rs10828317, odds ratio [OR] = 1.23; P = 2.30 × 10(-9)) and 10p14 marked by rs3824662 (OR = 1.31; P = 8.62 × 10(-12)). The single nucleotide polymorphism rs10828317 is responsible for the N215S polymorphism in exon 7 of PIP4K2A, and rs3824662 localizes to intron 3 of the transcription factor and putative tumor suppressor gene GATA3. The rs10828317 association was shown to be specifically associated with hyperdiploid ALL, whereas the rs3824662-associated risk was confined to nonhyperdiploid non-TEL-AML1 + ALL. The risk allele of rs3824662 was correlated with older age at diagnosis (P < .001) and significantly worse event-free survivorship (P < .0001). These findings provide further insights into the genetic and biological basis of inherited genetic susceptibility to BCP-ALL and the influence of constitutional genotype on disease development.


Subject(s)
Chromosomes, Human, Pair 10 , GATA3 Transcription Factor/genetics , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymorphism, Single Nucleotide , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Age Factors , Alleles , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Child , Exons , Female , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Introns , Male , Middle Aged , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Survival Analysis
16.
Nat Rev Cancer ; 6(3): 193-203, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16467884

ABSTRACT

Childhood leukaemia is the principal subtype of paediatric cancer and, despite success in treatment, its causes remain enigmatic. A plethora of candidate environmental exposures have been proposed, but most lack a biological rationale or consistent epidemiological evidence. Although there might not be a single or exclusive cause, an abnormal immune response to common infection(s) has emerged as a plausible aetiological mechanism.


Subject(s)
Antibody Formation/physiology , Infections/immunology , Leukemia/etiology , Child , Humans , Immunity, Cellular
17.
Lancet ; 381(9881): 1943-55, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23523389

ABSTRACT

Acute lymphoblastic leukaemia occurs in both children and adults but its incidence peaks between 2 and 5 years of age. Causation is multifactorial and exogenous or endogenous exposures, genetic susceptibility, and chance have roles. Survival in paediatric acute lymphoblastic leukaemia has improved to roughly 90% in trials with risk stratification by biological features of leukaemic cells and response to treatment, treatment modification based on patients' pharmacodynamics and pharmacogenomics, and improved supportive care. However, innovative approaches are needed to further improve survival while reducing adverse effects. Prognosis remains poor in infants and adults. Genome-wide profiling of germline and leukaemic cell DNA has identified novel submicroscopic structural genetic changes and sequence mutations that contribute to leukaemogenesis, define new disease subtypes, affect responsiveness to treatment, and might provide novel prognostic markers and therapeutic targets for personalised medicine.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Antineoplastic Agents/therapeutic use , Cell- and Tissue-Based Therapy/methods , Central Nervous System Diseases/therapy , Child , Child, Preschool , Chromosome Aberrations , Female , Forecasting , Gene Expression Profiling/methods , Gene Frequency , Gene Rearrangement, B-Lymphocyte , Genetic Predisposition to Disease/genetics , Genome, Human , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Recurrence , Remission Induction , Risk Assessment , Sequence Analysis, DNA , Transcription Factors/genetics , Translocation, Genetic/genetics , Treatment Outcome , Young Adult
18.
Proc Biol Sci ; 281(1781): 20132955, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24573849

ABSTRACT

Melanin provides a crucial filter for solar UV radiation and its genetically determined variation influences both skin pigmentation and risk of cancer. Genetic evidence suggests that the acquisition of a highly stable melanocortin 1 receptor allele promoting black pigmentation arose around the time of savannah colonization by hominins at some 1-2 Ma. The adaptive significance of dark skin is generally believed to be protection from UV damage but the pathologies that might have had a deleterious impact on survival and/or reproductive fitness, though much debated, are uncertain. Here, I suggest that data on age-associated cancer incidence and lethality in albinos living at low latitudes in both Africa and Central America support the contention that skin cancer could have provided a potent selective force for the emergence of black skin in early hominins.


Subject(s)
Albinism/physiopathology , Biological Evolution , Hominidae/genetics , Melanins/metabolism , Selection, Genetic , Skin Neoplasms/genetics , Skin Pigmentation/genetics , Animals , Hominidae/physiology , Melanins/biosynthesis , Melanins/genetics , Receptor, Melanocortin, Type 1/genetics , Skin Pigmentation/physiology
19.
Genes Chromosomes Cancer ; 52(10): 954-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893660

ABSTRACT

Over 90% of infants (< 1-year-old) diagnosed with leukemia have pro-B acute lymphoblastic leukemia (ALL) containing the MLL-AF4 fusion. When compared with other forms of paediatric ALL affecting later B-cell differentiation, MLL-AF4 pro-B is associated with a dismal prognosis with a typical 5-year disease-free survival of <20%. MLL-AF4 may be sufficient on its own for leukemogenesis or the gene-fusion product may alternatively predispose transformed cells to global genetic instability, enhancing the acquisition of additional key mutations. To gain insight into the genomic landscape of infant MLL-AF4 pro-B ALL we performed whole genome sequencing of diagnostic leukemic blasts and matched germline samples from three MLL-AF4 pro-B ALL infants. Our analysis revealed few somatic changes (copy number abnormalities, loss of heterozygosity, or single nucleotide variants), demonstrating that only a very small number of mutations are necessary to generate infant MLL-leukemia.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , DNA Copy Number Variations , DNA Mutational Analysis , Genome, Human , Genomics , Humans , INDEL Mutation/genetics , Infant , Infant, Newborn , Loss of Heterozygosity , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Translocation, Genetic
20.
Blood ; 118(18): 4910-8, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21900195

ABSTRACT

ETV6-RUNX1 gene fusion is usually an early, prenatal event in childhood acute lymphoblastic leukemia (ALL). Transformation results in the generation of a persistent (> 14 years) preleukemic clone, which postnatally converts to ALL after the acquisition of necessary secondary genetic alterations. Many cancer cells show some expression of the erythropoietin receptor (EPOR) gene, although the "functionality" of any EPOR complexes and their relevant signaling pathways in nonerythroid cells has not been validated. EPOR mRNA is selectively and ectopically expressed in ETV6-RUNX1(+) ALL, but the presence of a functional EPOR on the cell surface and its role in leukemogenesis driven by ETV6-RUNX1 remains to be identified. Here, we show that ETV6-RUNX1 directly binds the EPOR promoter and that expression of ETV6-RUNX1 alone in normal pre-B cells is sufficient to activate EPOR transcription. We further reveal that murine and human ETV6-RUNX1(+) cells expressing EPOR mRNA have EPO ligand binding activity that correlates with an increased cell survival through activation of the JAK2-STAT5 pathway and up-regulation of antiapoptotic BCL-XL. These data support the contention that ETV6-RUNX1 directly activates ectopic expression of a functional EPOR and provides cell survival signals that may contribute critically to persistence of covert premalignant clones in children.


Subject(s)
Oncogene Proteins, Fusion/physiology , Precursor Cells, B-Lymphoid/physiology , Receptors, Erythropoietin/physiology , Animals , Cell Lineage/genetics , Cell Lineage/physiology , Cell Survival/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit , Disease Models, Animal , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Precancerous Conditions/metabolism , Precursor Cells, B-Lymphoid/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL