Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Commun Med (Lond) ; 3(1): 109, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37567969

ABSTRACT

BACKGROUND: Xeroderma pigmentosum (XP) is a group of rare hereditary disorders with highly increased risk of skin tumors due to defective DNA repair. Recently we reported 34-fold increased risk of internal tumors in XP patients in comparison with general population. The molecular data and clinical practice on the internal tumors treatment in XP patients is limited and scarcely represented in the medical literature. In this work, we describe young patients with constitutive biallelic deactivation of the XPC gene developing gynecological tumors with somatic DICER1 mutations. METHODS: Whole genome sequencing was used to analyze in detail somatic mutational landscape and driver events of these rare tumors. RESULTS: We describe five early-onset gynecological tumors in four xeroderma pigmentosum group C (XP-C) young patients (11 to 19 years old) including vaginal embryonal rhabdomyosarcomas in monozygotic twin sisters, juvenile granulosa-cell tumor of the ovary and poorly differentiated stage IA Sertoli-Leydig cell tumor in 19-years old patient, and FIGO stage IC1 tumor of ovary in 13-years old patient. XP-C ovarian tumors harbor 4.4 times more single base substitutions than sporadic tissue-matched cancers and demonstrate XP-C specific mutation signature with strong transcriptional bias indicating inability of the cells to repair bulky DNA lesions of unknown etiology. A special mode of treatment was applied to avoid usage of chemotherapy which is toxic for XP patients. CONCLUSIONS: XP-C status should be accounted for prevention and specific treatment of gynecological tumors in young DNA repair-deficient XP patients.


Xeroderma pigmentosum group C (XP-C) is a rare inherited disorder resulting in a highly increased risk of skin and internal cancers due to the inability to efficiently repair DNA. In this study, we described four young XP-C patients who developed early-onset tumors affecting the female reproductive organs. We describe how we cared for these patients in the clinic. We looked at the genetic material within the tumors to better understand the mechanisms through which these tumors developed. We observed high numbers of specific types of changes in DNA, which are not typical for sporadic (non-inherited) gynecological tumors, but are characteristic of internal XP-C tumors. Further studies are needed to better understand the nature of these changes. Our findings highlight the important role of DNA repair in human tissues and cancer risk, and might inform future strategies for tumor prevention in XP-C patients.

2.
Cancer Discov ; 13(9): 1998-2011, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37377403

ABSTRACT

Several fibroblast growth factor receptor (FGFR) inhibitors are approved or in clinical development for the treatment of FGFR-driven urothelial cancer, and molecular mechanisms of resistance leading to patient relapses have not been fully explored. We identified 21 patients with FGFR-driven urothelial cancer treated with selective FGFR inhibitors and analyzed postprogression tissue and/or circulating tumor DNA (ctDNA). We detected single mutations in the FGFR tyrosine kinase domain in seven (33%) patients (FGFR3 N540K, V553L/M, V555L/M, E587Q; FGFR2 L551F) and multiple mutations in one (5%) case (FGFR3 N540K, V555L, and L608V). Using Ba/F3 cells, we defined their spectrum of resistance/sensitivity to multiple selective FGFR inhibitors. Eleven (52%) patients harbored alterations in the PI3K-mTOR pathway (n = 4 TSC1/2, n = 4 PIK3CA, n = 1 TSC1 and PIK3CA, n = 1 NF2, n = 1 PTEN). In patient-derived models, erdafitinib was synergistic with pictilisib in the presence of PIK3CA E545K, whereas erdafitinib-gefitinib combination was able to overcome bypass resistance mediated by EGFR activation. SIGNIFICANCE: In the largest study on the topic thus far, we detected a high frequency of FGFR kinase domain mutations responsible for resistance to FGFR inhibitors in urothelial cancer. Off-target resistance mechanisms involved primarily the PI3K-mTOR pathway. Our findings provide preclinical evidence sustaining combinatorial treatment strategies to overcome bypass resistance. See related commentary by Tripathi et al., p. 1964. This article is featured in Selected Articles from This Issue, p. 1949.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Neoplasm Recurrence, Local/drug therapy , Urinary Bladder Neoplasms/drug therapy , Carcinoma, Transitional Cell/drug therapy , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases , Class I Phosphatidylinositol 3-Kinases , Phosphatidylinositol 3-Kinases
3.
Nat Commun ; 14(1): 2561, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142601

ABSTRACT

Xeroderma pigmentosum (XP) is a genetic disorder caused by mutations in genes of the Nucleotide Excision Repair (NER) pathway (groups A-G) or in Translesion Synthesis DNA polymerase η (V). XP is associated with an increased skin cancer risk, reaching, for some groups, several thousand-fold compared to the general population. Here, we analyze 38 skin cancer genomes from five XP groups. We find that the activity of NER determines heterogeneity of the mutation rates across skin cancer genomes and that transcription-coupled NER extends beyond the gene boundaries reducing the intergenic mutation rate. Mutational profile in XP-V tumors and experiments with POLH knockout cell line reveal the role of polymerase η in the error-free bypass of (i) rare TpG and TpA DNA lesions, (ii) 3' nucleotides in pyrimidine dimers, and (iii) TpT photodimers. Our study unravels the genetic basis of skin cancer risk in XP and provides insights into the mechanisms reducing UV-induced mutagenesis in the general population.


Subject(s)
Skin Neoplasms , Xeroderma Pigmentosum , Humans , Xeroderma Pigmentosum/pathology , Ultraviolet Rays/adverse effects , DNA Repair/genetics , Mutation , Skin Neoplasms/genetics , Genomics
4.
Cancer Discov ; 13(5): 1116-1143, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36862804

ABSTRACT

Metastatic relapse after treatment is the leading cause of cancer mortality, and known resistance mechanisms are missing for most treatments administered to patients. To bridge this gap, we analyze a pan-cancer cohort (META-PRISM) of 1,031 refractory metastatic tumors profiled via whole-exome and transcriptome sequencing. META-PRISM tumors, particularly prostate, bladder, and pancreatic types, displayed the most transformed genomes compared with primary untreated tumors. Standard-of-care resistance biomarkers were identified only in lung and colon cancers-9.6% of META-PRISM tumors, indicating that too few resistance mechanisms have received clinical validation. In contrast, we verified the enrichment of multiple investigational and hypothetical resistance mechanisms in treated compared with nontreated patients, thereby confirming their putative role in treatment resistance. Additionally, we demonstrated that molecular markers improve 6-month survival prediction, particularly in patients with advanced breast cancer. Our analysis establishes the utility of the META-PRISM cohort for investigating resistance mechanisms and performing predictive analyses in cancer. SIGNIFICANCE: This study highlights the paucity of standard-of-care markers that explain treatment resistance and the promise of investigational and hypothetical markers awaiting further validation. It also demonstrates the utility of molecular profiling in advanced-stage cancers, particularly breast cancer, to improve the survival prediction and assess eligibility to phase I clinical trials. This article is highlighted in the In This Issue feature, p. 1027.


Subject(s)
Breast Neoplasms , Neoplasms, Second Primary , Male , Humans , Transcriptome , Neoplasm Recurrence, Local , Breast Neoplasms/drug therapy , Genomics , Gene Expression Profiling
5.
Cancer Discov ; 13(4): 858-879, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36669143

ABSTRACT

Cancer immunotherapy combinations have recently been shown to improve the overall survival of advanced mesotheliomas, especially for patients responding to those treatments. We aimed to characterize the biological correlates of malignant pleural mesotheliomas' primary resistance to immunotherapy and antiangiogenics by testing the combination of pembrolizumab, an anti-PD-1 antibody, and nintedanib, a pan-antiangiogenic tyrosine kinase inhibitor, in the multicenter PEMBIB trial (NCT02856425). Thirty patients with advanced malignant pleural mesothelioma were treated and explored. Unexpectedly, we found that refractory patients were actively recruiting CD3+CD8+ cytotoxic T cells in their tumors through CXCL9 tumor release upon treatment. However, these patients displayed high levels of somatic copy-number alterations in their tumors that correlated with high blood and tumor levels of IL6 and CXCL8. Those proinflammatory cytokines resulted in higher tumor secretion of VEGF and tumor enrichment in regulatory T cells. Advanced mesothelioma should further benefit from stratified combination therapies adapted to their tumor biology. SIGNIFICANCE: Sequential explorations of fresh tumor biopsies demonstrated that mesothelioma resistance to anti-PD-1 + antiangiogenics is not due to a lack of tumor T-cell infiltration but rather due to adaptive immunosuppressive pathways by tumors, involving molecules (e.g., IL6, CXCL8, VEGF, and CTLA4) that are amenable to targeted therapies. This article is highlighted in the In This Issue feature, p. 799.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Humans , Interleukin-6 , Vascular Endothelial Growth Factor A , Lung Neoplasms/genetics , Mesothelioma/drug therapy , Mesothelioma/genetics , Immunotherapy , Genomic Instability , Inflammation/drug therapy , Inflammation/genetics , Pleural Neoplasms/drug therapy , Pleural Neoplasms/genetics
6.
Clin Cancer Res ; 28(7): 1422-1432, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35078858

ABSTRACT

PURPOSE: Vismodegib is approved for the treatment of locally advanced basal cell carcinoma (laBCC), but some cases demonstrate intrinsic resistance (IR) to the drug. We sought to assess the frequency of IR to vismodegib in laBCC and its underlying genomic mechanisms. EXPERIMENTAL DESIGN: Response to vismodegib was evaluated in a cohort of 148 laBCC patients. Comprehensive genomic and transcriptomic profiling was performed in a subset of five intrinsically resistant BCC (IR-BCC). RESULTS: We identified that IR-BCC represents 6.1% of laBCC in the studied cohort. Prior treatment with chemotherapy was associated with IR. Genetic events that were previously associated with acquired resistance (AR) in BCC or medulloblastoma were observed in three out of five IR-BCC. However, IR-BCCs were distinct by highly rearranged polyploid genomes. Functional analyses identified hyperactivation of the HIPPO-YAP and WNT pathways at RNA and protein levels in IR-BCC. In vitro assay on the BCC cell line further confirmed that YAP1 overexpression increases the cell proliferation rate. CONCLUSIONS: IR to vismodegib is a rare event in laBCC. IR-BCCs frequently harbor resistance mutations in the Hh pathway, but also are characterized by hyperactivation of the HIPPO-YAP and WNT pathways.


Subject(s)
Antineoplastic Agents , Carcinoma, Basal Cell , Cerebellar Neoplasms , Skin Neoplasms , Anilides/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cerebellar Neoplasms/drug therapy , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Pyridines , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology
7.
Nat Genet ; 51(1): 36-41, 2019 01.
Article in English | MEDLINE | ID: mdl-30510240

ABSTRACT

Studies in experimental systems have identified a multitude of mutational mechanisms including DNA replication infidelity and DNA damage followed by inefficient repair or replicative bypass. However, the relative contributions of these mechanisms to human germline mutation remain unknown. Here, we show that error-prone damage bypass on the lagging strand plays a major role in human mutagenesis. Transcription-coupled DNA repair removes lesions on the transcribed strand; lesions on the non-transcribed strand are preferentially converted into mutations. In human polymorphism we detect a striking similarity between mutation types predominant on the non-transcribed strand and on the strand lagging during replication. Moreover, damage-induced mutations in cancers accumulate asymmetrically with respect to the direction of replication, suggesting that DNA lesions are resolved asymmetrically. We experimentally demonstrate that replication delay greatly attenuates the mutagenic effect of ultraviolet irradiation, confirming that replication converts DNA damage into mutations. We estimate that at least 10% of human mutations arise due to DNA damage.


Subject(s)
DNA Replication/genetics , DNA/genetics , Germ-Line Mutation/genetics , Neoplasms/genetics , Cells, Cultured , DNA Damage/genetics , DNA Repair/genetics , Humans , Mutagenesis/genetics , Polymorphism, Single Nucleotide/genetics , Transcription, Genetic/genetics
8.
Oncoimmunology ; 7(12): e1501137, 2018.
Article in English | MEDLINE | ID: mdl-30524896

ABSTRACT

Immune checkpoint blockade (ICB) is currently evaluated in patients with glioblastoma (GBM), based on encouraging clinical data in other cancers, and results from studies with the methylcholanthrene-induced GL261 mouse glioma. In this paper, we describe a novel model faithfully recapitulating some key human GBM characteristics, including low mutational load, a factor reported as a prognostic indicator of ICB response. Consistent with this observation, SB28 is completely resistant to ICB, contrasting with treatment sensitivity of the more highly mutated GL261. Moreover, SB28 shows features of a poorly immunogenic tumor, with low MHC-I expression and modest CD8+ T-cell infiltration, suggesting that it may present similar challenges for immunotherapy as human GBM. Based on these key features for immune reactivity, SB28 may represent a treatment-resistant malignancy likely to mirror responses of many human tumors. We therefore propose that SB28 is a particularly suitable model for optimization of GBM immunotherapy.

10.
N Engl J Med ; 378(3): 250-261, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29298116

ABSTRACT

BACKGROUND: Sporadic arteriovenous malformations of the brain, which are morphologically abnormal connections between arteries and veins in the brain vasculature, are a leading cause of hemorrhagic stroke in young adults and children. The genetic cause of this rare focal disorder is unknown. METHODS: We analyzed tissue and blood samples from patients with arteriovenous malformations of the brain to detect somatic mutations. We performed exome DNA sequencing of tissue samples of arteriovenous malformations of the brain from 26 patients in the main study group and of paired blood samples from 17 of those patients. To confirm our findings, we performed droplet digital polymerase-chain-reaction (PCR) analysis of tissue samples from 39 patients in the main study group (21 with matching blood samples) and from 33 patients in an independent validation group. We interrogated the downstream signaling pathways, changes in gene expression, and cellular phenotype that were induced by activating KRAS mutations, which we had discovered in tissue samples. RESULTS: We detected somatic activating KRAS mutations in tissue samples from 45 of the 72 patients and in none of the 21 paired blood samples. In endothelial cell-enriched cultures derived from arteriovenous malformations of the brain, we detected KRAS mutations and observed that expression of mutant KRAS (KRASG12V) in endothelial cells in vitro induced increased ERK (extracellular signal-regulated kinase) activity, increased expression of genes related to angiogenesis and Notch signaling, and enhanced migratory behavior. These processes were reversed by inhibition of MAPK (mitogen-activated protein kinase)-ERK signaling. CONCLUSIONS: We identified activating KRAS mutations in the majority of tissue samples of arteriovenous malformations of the brain that we analyzed. We propose that these malformations develop as a result of KRAS-induced activation of the MAPK-ERK signaling pathway in brain endothelial cells. (Funded by the Swiss Cancer League and others.).


Subject(s)
Intracranial Arteriovenous Malformations/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Cells, Cultured , DNA Mutational Analysis , Exome , Gene Expression , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracranial Arteriovenous Malformations/etiology , Intracranial Arteriovenous Malformations/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism
11.
J Pathol ; 243(3): 331-341, 2017 11.
Article in English | MEDLINE | ID: mdl-28805995

ABSTRACT

Biallelic mismatch repair deficiency (bMMRD) in tumours is frequently associated with somatic mutations in the exonuclease domains of DNA polymerases POLE or POLD1, and results in a characteristic mutational profile. In this article, we describe the genetic basis of ultramutated high-grade brain tumours in the context of bMMRD. We performed exome sequencing of two second-cousin patients from a large consanguineous family of Indian origin with early onset of high-grade glioblastoma and astrocytoma. We identified a germline homozygous nonsense variant, p.R802*, in the PMS2 gene. Additionally, by genome sequencing of these tumours, we found extremely high somatic mutation rates (237/Mb and 123/Mb), as well as somatic mutations in the proofreading domain of POLE polymerase (p.P436H and p.L424V), which replicates the leading DNA strand. Most interestingly, we found, in both cancers, that the vast majority of mutations were consistent with the signature of POLE exo- , i.e. an abundance of C>A and C>T mutations, particularly in special contexts, on the leading strand. We showed that the fraction of mutations under positive selection among mutations in tumour suppressor genes is more than two-fold lower in ultramutated tumours than in other glioblastomas. Genetic analyses enabled the diagnosis of the two consanguineous childhood brain tumours as being due to a combination of PMS2 germline and POLE somatic variants, and confirmed them as bMMRD/POLE exo- disorders. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Brain Neoplasms/genetics , DNA Mismatch Repair/genetics , DNA Polymerase II/genetics , Genetic Predisposition to Disease , Germ-Line Mutation/genetics , Mismatch Repair Endonuclease PMS2/genetics , Brain Neoplasms/pathology , DNA/genetics , Female , Humans , Male , Poly-ADP-Ribose Binding Proteins
12.
Genome Res ; 27(8): 1336-1343, 2017 08.
Article in English | MEDLINE | ID: mdl-28512192

ABSTRACT

Mismatch repair (MMR) is one of the main systems maintaining fidelity of replication. Differences in correction of errors produced during replication of the leading and the lagging DNA strands were reported in yeast and in human cancers, but the causes of these differences remain unclear. Here, we analyze data on human cancers with somatic mutations in two of the major DNA polymerases, delta and epsilon, that replicate the genome. We show that these cancers demonstrate a substantial asymmetry of the mutations between the leading and the lagging strands. The direction of this asymmetry is the opposite between cancers with mutated polymerases delta and epsilon, consistent with the role of these polymerases in replication of the lagging and the leading strands in human cells, respectively. Moreover, the direction of strand asymmetry observed in cancers with mutated polymerase delta is similar to that observed in MMR-deficient cancers. Together, these data indicate that polymerase delta (possibly together with polymerase alpha) contributes more mismatches during replication than its leading-strand counterpart, polymerase epsilon; that most of these mismatches are repaired by the MMR system; and that MMR repairs about three times more mismatches produced in cells during lagging strand replication compared with the leading strand.


Subject(s)
DNA Mismatch Repair/genetics , DNA Polymerase III/genetics , DNA Polymerase II/genetics , DNA Replication , Mutation , Neoplasms/genetics , Exome , Humans , Mutation Rate , Whole Genome Sequencing
13.
Nat Genet ; 48(4): 398-406, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950094

ABSTRACT

Basal cell carcinoma (BCC) of the skin is the most common malignant neoplasm in humans. BCC is primarily driven by the Sonic Hedgehog (Hh) pathway. However, its phenotypic variation remains unexplained. Our genetic profiling of 293 BCCs found the highest mutation rate in cancer (65 mutations/Mb). Eighty-five percent of the BCCs harbored mutations in Hh pathway genes (PTCH1, 73% or SMO, 20% (P = 6.6 × 10(-8)) and SUFU, 8%) and in TP53 (61%). However, 85% of the BCCs also harbored additional driver mutations in other cancer-related genes. We observed recurrent mutations in MYCN (30%), PPP6C (15%), STK19 (10%), LATS1 (8%), ERBB2 (4%), PIK3CA (2%), and NRAS, KRAS or HRAS (2%), and loss-of-function and deleterious missense mutations were present in PTPN14 (23%), RB1 (8%) and FBXW7 (5%). Consistent with the mutational profiles, N-Myc and Hippo-YAP pathway target genes were upregulated. Functional analysis of the mutations in MYCN, PTPN14 and LATS1 suggested their potential relevance in BCC tumorigenesis.


Subject(s)
Carcinoma, Basal Cell/genetics , Signal Transduction/radiation effects , Skin Neoplasms/genetics , Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/pathology , DNA Mutational Analysis , Disease Progression , Exome , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mutation , Pyridines/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Transcriptome
14.
Genome Res ; 26(2): 174-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26755635

ABSTRACT

APOBEC3A and APOBEC3B, cytidine deaminases of the APOBEC family, are among the main factors causing mutations in human cancers. APOBEC deaminates cytosines in single-stranded DNA (ssDNA). A fraction of the APOBEC-induced mutations occur as clusters ("kataegis") in single-stranded DNA produced during repair of double-stranded breaks (DSBs). However, the properties of the remaining 87% of nonclustered APOBEC-induced mutations, the source and the genomic distribution of the ssDNA where they occur, are largely unknown. By analyzing genomic and exomic cancer databases, we show that >33% of dispersed APOBEC-induced mutations occur on the lagging strand during DNA replication, thus unraveling the major source of ssDNA targeted by APOBEC in cancer. Although methylated cytosine is generally more mutation-prone than nonmethylated cytosine, we report that methylation reduces the rate of APOBEC-induced mutations by a factor of roughly two. Finally, we show that in cancers with extensive APOBEC-induced mutagenesis, there is almost no increase in mutation rates in late replicating regions (contrary to other cancers). Because late-replicating regions are depleted in exons, this results in a 1.3-fold higher fraction of mutations residing within exons in such cancers. This study provides novel insight into the APOBEC-induced mutagenesis and describes the peculiarity of the mutational processes in cancers with the signature of APOBEC-induced mutations.


Subject(s)
Cytidine Deaminase/physiology , Neoplasms/genetics , Cytosine/metabolism , DNA Methylation , DNA Mutational Analysis , DNA Replication , Exome , Humans , Mutagenesis , Mutation , Mutation Rate
15.
Am J Hum Genet ; 95(6): 660-74, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25480033

ABSTRACT

Gene expression levels can be subject to selection. We hypothesized that the age of gene origin is associated with expression constraints, given that it affects the level of gene integration into the functional cellular environment. By studying the genetic variation affecting gene expression levels (cis expression quantitative trait loci [cis-eQTLs]) and protein levels (cis protein QTLs [cis-pQTLs]), we determined that young, primate-specific genes are enriched in cis-eQTLs and cis-pQTLs. Compared to cis-eQTLs of old genes originating before the zebrafish divergence, cis-eQTLs of young genes have a higher effect size, are located closer to the transcription start site, are more significant, and tend to influence genes in multiple tissues and populations. These results suggest that the expression constraint of each gene increases throughout its lifespan. We also detected a positive correlation between expression constraints (approximated by cis-eQTL properties) and coding constraints (approximated by Ka/Ks) and observed that this correlation might be driven by gene age. To uncover factors associated with the increase in gene-age-related expression constraints, we demonstrated that gene connectivity, gene involvement in complex regulatory networks, gene haploinsufficiency, and the strength of posttranscriptional regulation increase with gene age. We also observed an increase in heritability of gene expression levels with age, implying a reduction of the environmental component. In summary, we show that gene age shapes key gene properties during evolution and is therefore an important component of genome function.


Subject(s)
Gene Expression Regulation , Genetic Variation , Genome/genetics , Proteins/genetics , Quantitative Trait Loci/genetics , Age Factors , Cell Line , Female , Fetal Blood , Fibroblasts , Gene Expression Profiling , Humans , Infant, Newborn , Logistic Models , Male , Organ Specificity , Polymorphism, Single Nucleotide , Proteins/metabolism , Transcription Initiation Site , Umbilical Cord
16.
Nat Commun ; 5: 4654, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25105841

ABSTRACT

Children with Down syndrome (DS) and acute lymphoblastic leukaemia (ALL) have poorer survival and more relapses than non-DS children with ALL, highlighting an urgent need for deeper mechanistic understanding of DS-ALL. Here, using full-exome or cancer genes-targeted sequencing of 42 ALL samples from 39 DS patients, we uncover driver mutations in RAS, (KRAS and NRAS) recurring to a similar extent (15/42) as JAK2 (12/42) mutations or P2RY8-CRLF2 fusions (14/42). RAS mutations are almost completely mutually exclusive with JAK2 mutations (P=0.016), driving a combined total of two-thirds of analysed cases. Clonal architecture analysis reveals that both RAS and JAK2 drove sub-clonal expansions primarily initiated by CRLF2 rearrangements, and/or mutations in chromatin remodellers and lymphocyte differentiation factors. Remarkably, in 2/3 relapsed cases, there is a switch from a primary JAK2- or PTPN11-mutated sub-clone to a RAS-mutated sub-clone in relapse. These results provide important new insights informing the patient stratification strategies for targeted therapeutic approaches for DS-ALL.


Subject(s)
Down Syndrome/genetics , Janus Kinase 2/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , ras Proteins/metabolism , Child , Chromatin/chemistry , DNA Mutational Analysis , Down Syndrome/metabolism , Female , Gene Deletion , Gene Expression Regulation, Leukemic , Humans , Lymphocytes/cytology , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Cytokine/genetics
17.
Blood ; 122(4): 554-61, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23733339

ABSTRACT

Some neonates with Down syndrome (DS) are diagnosed with self-regressing transient myeloproliferative disorder (TMD), and 20% to 30% of those progress to acute megakaryoblastic leukemia (AMKL). We performed exome sequencing in 7 TMD/AMKL cases and copy-number analysis in these and 10 additional cases. All TMD/AMKL samples contained GATA1 mutations. No exome-sequenced TMD/AMKL sample had other recurrently mutated genes. However, 2 of 5 TMD cases, and all AMKL cases, showed mutations/deletions other than GATA1, in genes proven as transformation drivers in non-DS leukemia (EZH2, APC, FLT3, JAK1, PARK2-PACRG, EXT1, DLEC1, and SMC3). One patient at the TMD stage revealed 2 clonal expansions with different GATA1 mutations, of which 1 clone had an additional driver mutation. Interestingly, it was the other clone that gave rise to AMKL after accumulating mutations in 7 other genes. Data suggest that GATA1 mutations alone are sufficient for clonal expansions, and additional driver mutations at the TMD stage do not necessarily predict AMKL progression. Later in infancy, leukemic progression requires "third-hit driver" mutations/somatic copy-number alterations found in non-DS leukemias. Putative driver mutations affecting WNT (wingless-related integration site), JAK-STAT (Janus kinase/signal transducer and activator of transcription), or MAPK/PI3K (mitogen-activated kinase/phosphatidylinositol-3 kinase) pathways were found in all cases, aberrant activation of which converges on overexpression of MYC.


Subject(s)
Cell Transformation, Neoplastic/genetics , Down Syndrome/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Myeloproliferative Disorders/genetics , Disease Progression , Down Syndrome/complications , Exome/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genomic Instability/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Infant, Newborn , Leukemia, Megakaryoblastic, Acute/complications , Leukemia, Megakaryoblastic, Acute/pathology , Microarray Analysis , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/pathology , Polymorphism, Single Nucleotide , Transcriptome
18.
Cell Signal ; 25(4): 743-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23280189

ABSTRACT

The Ccr4-Not complex is a conserved multi-subunit complex in eukaryotes that carries 2 enzymatic activities: ubiquitination mediated by the Not4 RING E3 ligase and deadenylation mediated by the Ccr4 and Caf1 orthologs. This complex has been implicated in all aspects of the mRNA life cycle, from synthesis of mRNAs in the nucleus to their degradation in the cytoplasm. More recently the complex has also been implicated in many aspects of the life cycle of proteins, from quality control during synthesis of peptides, to assembly of protein complexes and protein degradation. Consistently, the Ccr4-Not complex is found both in the nucleus, where it is connected to transcribing ORFs, and in the cytoplasm, where it was revealed to be both associated with translating ribosomes and in RNA processing bodies. This functional and physical presence of the Ccr4-Not complex at all stages of gene expression raises the question of its fundamental role. This review will summarize recent evidence designing the Not3/5 module of the Ccr4-Not complex as a functional module involved in coordination of the regulation of gene expression between the nucleus and the cytoplasm.


Subject(s)
Receptors, CCR4/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Fungal , Humans , Protein Binding , RNA, Messenger/metabolism , Receptors, CCR4/chemistry , Receptors, CCR4/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
19.
Mol Biol Evol ; 30(2): 347-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22983951

ABSTRACT

The mammalian mitochondrial genomes differ from the nuclear genomes by maternal inheritance, absence of recombination, and higher mutation rate. All these differences decrease the effective population size of mitochondrial genome and make it more susceptible to accumulation of slightly deleterious mutations. It was hypothesized that mitochondrial genes, especially in species with low effective population size, irreversibly degrade leading to decrease of organismal fitness and even to extinction of species through the mutational meltdown. To interrogate this hypothesis, we compared the purifying selections acting on the representative set of mitochondrial (potentially degrading) and nuclear (potentially not degrading) protein-coding genes in species with different effective population size. For 21 mammalian species, we calculated the ratios of accumulation of slightly deleterious mutations approximated by Kn/Ks separately for mitochondrial and nuclear genomes. The 75% of variation in Kn/Ks is explained by two independent variables: type of a genome (mitochondrial or nuclear) and effective population size of species approximated by generation time. First, we observed that purifying selection is more effective in mitochondria than in the nucleus that implies strong evolutionary constraints of mitochondrial genome. Mitochondrial de novo nonsynonymous mutations have at least 5-fold more harmful effect when compared with nuclear. Second, Kn/Ks of mitochondrial and nuclear genomes is positively correlated with generation time of species, indicating relaxation of purifying selection with decrease of species-specific effective population size. Most importantly, the linear regression lines of mitochondrial and nuclear Kn/Ks's from generation times of species are parallel, indicating congruent relaxation of purifying selection in both genomes. Thus, our results reveal that the distribution of selection coefficients of de novo nonsynonymous mitochondrial mutations has a similar shape with the distribution of de novo nonsynonymous nuclear mutations, but its mean is five times smaller. The harmful effect of mitochondrial de novo nonsynonymous mutations triggers highly effective purifying selection, which maintains the fitness of the mammalian mitochondrial genome.


Subject(s)
Evolution, Molecular , Genome, Mitochondrial , Genome , Mitochondrial Proteins/genetics , Selection, Genetic , Amino Acid Substitution , Animals , Cell Nucleus/genetics , Female , Humans , Male , Mammals/genetics , Mutation
20.
Cancer Res ; 72(23): 6279-89, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23204322

ABSTRACT

Oncogene-induced DNA replication stress is thought to drive genomic instability in cancer. In particular, replication stress can explain the high prevalence of focal genomic deletions mapping within very large genes in human tumors. However, the origin of single-nucleotide substitutions (SNS) in nonfamilial cancers is strongly debated. Some argue that cancers have a mutator phenotype, whereas others argue that the normal DNA replication error rates are sufficient to explain the number of observed SNSs. Here, we sequenced the exomes of 24, mostly precancerous, colon polyps. Analysis of the sequences revealed mutations in the APC, CTNNB1, and BRAF genes as the presumptive cancer-initiating events and many passenger SNSs. We used the number of SNSs in the various lesions to calculate mutation rates for normal colon and adenomas and found that colon adenomas exhibit a mutator phenotype. Interestingly, the SNSs in the adenomas mapped more often than expected within very large genes, where focal deletions in response to DNA replication stress also map. We propose that single-stranded DNA generated in response to oncogene-induced replication stress compromises the repair of deaminated cytosines and other damaged bases, leading to the observed SNS mutator phenotype.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Exome , Adenoma/pathology , Colorectal Neoplasms/pathology , DNA Repair/genetics , Genome, Human , Genomic Instability , Humans , Mutation , Phenotype , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL