Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Adv Exp Med Biol ; 1361: 75-100, 2022.
Article in English | MEDLINE | ID: mdl-35230684

ABSTRACT

Microsatellite instability (MSI) is a genetic alteration due to a deficiency of the DNA mismatch repair system, where microsatellites accumulate insertions/deletions. This phenotype has been extensively characterized in colorectal cancer and is also sought in the context of Lynch syndrome diagnosis. It has recently been described in dozens of cancer types from whole genome/exome sequencing data, bearing some prognostic information. Moreover, MSI has also proven to be a major predicator of the response to immune checkpoint blockade therapy in solid cancer patients. Among the different methods developed for MSI detection in cancer, next-generation sequencing (NGS) is a promising and versatile technology offering many possibilities and advantages in diverse clinical applications compared to the gold standard PCR and capillary electrophoresis approach. NGS could notably increase the number of analyzed microsatellites and potentially be used to analyze other genetic alterations required for precision oncology. However, it requires the development of robust new computational algorithms for the analysis of NGS microsatellite data. In this chapter, we describe the different approaches developed for the assessment of MSI from NGS data in cancer, including the different microsatellite panels and computational algorithms proposed, highlighting their advantages and drawbacks, and their evaluation in different clinical applications.


Subject(s)
Colorectal Neoplasms , Neoplasms , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Mismatch Repair , High-Throughput Nucleotide Sequencing/methods , Humans , Microsatellite Instability , Microsatellite Repeats/genetics , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine
2.
Nat Commun ; 9(1): 5235, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531861

ABSTRACT

Cyclins A2 and E1 regulate the cell cycle by promoting S phase entry and progression. Here, we identify a hepatocellular carcinoma (HCC) subgroup exhibiting cyclin activation through various mechanisms including hepatitis B virus (HBV) and adeno-associated virus type 2 (AAV2) insertions, enhancer hijacking and recurrent CCNA2 fusions. Cyclin A2 or E1 alterations define a homogenous entity of aggressive HCC, mostly developed in non-cirrhotic patients, characterized by a transcriptional activation of E2F and ATR pathways and a high frequency of RB1 and PTEN inactivation. Cyclin-driven HCC display a unique signature of structural rearrangements with hundreds of tandem duplications and templated insertions frequently activating TERT promoter. These rearrangements, strongly enriched in early-replicated active chromatin regions, are consistent with a break-induced replication mechanism. Pan-cancer analysis reveals a similar signature in BRCA1-mutated breast and ovarian cancers. Together, this analysis reveals a new poor prognosis HCC entity and a rearrangement signature related to replication stress.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclin A2/genetics , Cyclin E/genetics , Gene Rearrangement , Liver Neoplasms/genetics , Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cyclin A2/metabolism , Cyclin E/metabolism , Dependovirus , Female , Gene Expression Regulation, Neoplastic , Hepatitis B virus/genetics , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , Mutagenesis, Insertional , Oncogene Proteins/metabolism , Parvovirinae/genetics , Promoter Regions, Genetic/genetics , Survival Analysis
3.
Nat Commun ; 8(1): 1315, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29101368

ABSTRACT

Genomic alterations driving tumorigenesis result from the interaction of environmental exposures and endogenous cellular processes. With a diversity of risk factors, liver cancer is an ideal model to study these interactions. Here, we analyze the whole genomes of 44 new and 264 published liver cancers and we identify 10 mutational and 6 structural rearrangement signatures showing distinct relationships with environmental exposures, replication, transcription, and driver genes. The liver cancer-specific signature 16, associated with alcohol, displays a unique feature of transcription-coupled damage and is the main source of CTNNB1 mutations. Flood of insertions/deletions (indels) are identified in very highly expressed hepato-specific genes, likely resulting from replication-transcription collisions. Reconstruction of sub-clonal architecture reveals mutational signature evolution during tumor development exemplified by the vanishing of aflatoxin B1 signature in African migrants. Finally, chromosome duplications occur late and may represent rate-limiting events in tumorigenesis. These findings shed new light on the natural history of liver cancers.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Mutation , Carcinoma, Hepatocellular/etiology , Chromosome Duplication , DNA Mutational Analysis , DNA Replication , Evolution, Molecular , Female , Gene Dosage , Gene Rearrangement , Genome, Human , Humans , INDEL Mutation , Liver Neoplasms/etiology , Male , Risk Factors , Transcription, Genetic , Whole Genome Sequencing
4.
Mol Oncol ; 11(11): 1561-1578, 2017 11.
Article in English | MEDLINE | ID: mdl-28815885

ABSTRACT

Immune checkpoints are powerful inhibitory molecules that promote tumor survival. Their blockade is now recognized as providing effective therapeutic benefit against cancer. Human leukocyte antigen G (HLA-G), a recently identified immune checkpoint, has been detected in many types of primary tumors and metastases, in malignant effusions as well as on tumor-infiltrating cells, particularly in patients with clear cell renal cell carcinoma (ccRCC). Here, in order to define a possible anticancer therapy, we used a molecular approach based on an unbiased strategy that combines transcriptome determination and immunohistochemical labeling, to analyze in-depth the HLA-G isoforms expressed in these tumors. We found that the expression of HLA-G is highly variable among tumors and distinct areas of the same tumor, testifying a marked inter- and intratumor heterogeneity. Moreover, our results generate an inventory of novel HLA-G isoforms which includes spliced forms that have an extended 5'-region and lack the transmembrane and alpha-1 domains. So far, these isoforms could not be detected by any method available and their assessment may improve the procedure by which tumors are analyzed. Collectively, our approach provides the first extensive portrait of HLA-G in ccRCC and reveals data that should prove suitable for the tailoring of future clinical applications.


Subject(s)
Carcinoma, Renal Cell/genetics , HLA-G Antigens/genetics , Kidney Neoplasms/genetics , Transcriptome , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , HLA-G Antigens/analysis , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/pathology , Protein Isoforms/analysis , Protein Isoforms/genetics , Sequence Analysis, RNA
5.
Nat Commun ; 5: 5135, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25351205

ABSTRACT

The incidence of renal cell carcinoma (RCC) is increasing worldwide, and its prevalence is particularly high in some parts of Central Europe. Here we undertake whole-genome and transcriptome sequencing of clear cell RCC (ccRCC), the most common form of the disease, in patients from four different European countries with contrasting disease incidence to explore the underlying genomic architecture of RCC. Our findings support previous reports on frequent aberrations in the epigenetic machinery and PI3K/mTOR signalling, and uncover novel pathways and genes affected by recurrent mutations and abnormal transcriptome patterns including focal adhesion, components of extracellular matrix (ECM) and genes encoding FAT cadherins. Furthermore, a large majority of patients from Romania have an unexpected high frequency of A:T>T:A transversions, consistent with exposure to aristolochic acid (AA). These results show that the processes underlying ccRCC tumorigenesis may vary in different populations and suggest that AA may be an important ccRCC carcinogen in Romania, a finding with major public health implications.


Subject(s)
Carcinoma, Renal Cell/genetics , Genetic Variation , Genome, Human/genetics , Genomics , Adult , Aged , Aged, 80 and over , Cohort Studies , Europe , Female , Focal Adhesions/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mutation , Mutation Rate , Oncogene Proteins, Fusion/genetics , Phosphatidylinositol 3-Kinases/genetics , RNA Splicing/genetics , Sequence Analysis, DNA , Signal Transduction/genetics
6.
Nat Med ; 17(10): 1283-9, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21946539

ABSTRACT

Heat shock proteins (HSPs) are necessary for cancer cell survival. We identified a mutant of HSP110 (HSP110ΔE9) in colorectal cancer showing microsatellite instability (MSI CRC), generated from an aberrantly spliced mRNA and lacking the HSP110 substrate-binding domain. This mutant was expressed at variable levels in almost all MSI CRC cell lines and primary tumors tested. HSP110ΔE9 impaired both the normal cellular localization of HSP110 and its interaction with other HSPs, thus abrogating the chaperone activity and antiapoptotic function of HSP110 in a dominant-negative manner. HSP110ΔE9 overexpression caused the sensitization of cells to anticancer agents such as oxaliplatin and 5-fluorouracil, which are routinely prescribed in the adjuvant treatment of people with CRC. The survival and response to chemotherapy of subjects with MSI CRCs was associated with the tumor expression level of HSP110ΔE9. HSP110 may thus constitute a major determinant for both prognosis and treatment response in CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , HSP110 Heat-Shock Proteins/metabolism , Antineoplastic Agents/therapeutic use , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Primers/genetics , Fluorescent Antibody Technique , Fluorouracil , HSP110 Heat-Shock Proteins/genetics , Humans , Immunoprecipitation , Microsatellite Instability , Mutation/genetics , Organoplatinum Compounds , Oxaliplatin , Plasmids/genetics , Prognosis , Real-Time Polymerase Chain Reaction , Regression Analysis , Transfection
7.
Science ; 332(6026): 240-3, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474761

ABSTRACT

The spliceosome, a ribonucleoprotein complex that includes proteins and small nuclear RNAs (snRNAs), catalyzes RNA splicing through intron excision and exon ligation to produce mature messenger RNAs, which, in turn serve as templates for protein translation. We identified four point mutations in the U4atac snRNA component of the minor spliceosome in patients with brain and bone malformations and unexplained postnatal death [microcephalic osteodysplastic primordial dwarfism type 1 (MOPD 1) or Taybi-Linder syndrome (TALS); Mendelian Inheritance in Man ID no. 210710]. Expression of a subgroup of genes, possibly linked to the disease phenotype, and minor intron splicing were affected in cell lines derived from TALS patients. Our findings demonstrate a crucial role of the minor spliceosome component U4atac snRNA in early human development and postnatal survival.


Subject(s)
Point Mutation , RNA Splicing , RNA, Small Nuclear/genetics , Spliceosomes/genetics , Base Pairing , Cell Line , Child, Preschool , Chromosomes, Human, Pair 2/genetics , Dwarfism/genetics , Dwarfism/metabolism , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Humans , Infant , Introns , Inverted Repeat Sequences , Male , Microcephaly/genetics , Microcephaly/metabolism , Microtubule-Associated Proteins/genetics , Nucleic Acid Conformation , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Pedigree , RNA Splice Sites , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Spliceosomes/metabolism
8.
J Natl Cancer Inst ; 102(22): 1731-40, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-20923998

ABSTRACT

BACKGROUND: The thiopurine prodrug azathioprine is used extensively in cancer therapy. Exposure to this drug results in the selection of DNA mismatch repair-deficient cell clones in vitro. It has also been suggested that thiopurine drugs might constitute a risk factor for the emergence of human neoplasms displaying microsatellite instability (MSI) because of deficient DNA mismatch repair. METHODS: Azathioprine was administered via drinking water (6-20 mg/kg body weight per day) to mice that were null (Msh2⁻(/)⁻; n = 27), heterozygous (Msh2(+/)⁻; n = 22), or wild type (Msh2(WT); n = 18) for the DNA mismatch repair gene Msh2. Control mice (45 Msh2⁻(/)⁻, 38 Msh2(+/)⁻, and 12 Msh2(WT)) received drinking water lacking azathioprine. The effect of azathioprine on tumorigenesis and survival of the mice was evaluated by Kaplan-Meier curves using log-rank and Gehan-Breslow-Wilcoxon tests. Mouse tumor samples were characterized by histology and immunophenotyping, and their MSI status was determined by polymerase chain reaction analysis of three noncoding microsatellite markers and by immunohistochemistry. Msh2 status of tumor samples was assessed by loss of heterozygosity analyses and sequencing after reverse transcription-polymerase chain reaction of the entire Msh2 coding sequence. All statistical tests were two-sided. RESULTS: Most untreated Msh2(WT) and Msh2(+/)⁻ mice remained asymptomatic and alive at 250 days of age, whereas azathioprine-treated Msh2(WT) and Msh2(+/)⁻ mice developed lymphomas and died prematurely (median survival of 71 and 165 days of age, respectively). Azathioprine-treated Msh2(+/)⁻ mice developed diffuse lymphomas lacking Msh2 expression and displaying MSI due to somatic inactivation of the functional Msh2 allele by loss of heterozygosity or mutation. By contrast, azathioprine-treated Msh2(WT) mice displayed no obvious tumor phenotype, but histological examination showed microscopic splenic foci of neoplastic lymphoid cells that retained Msh2 expression and did not display MSI. Both untreated and azathioprine-treated Msh2⁻(/)⁻ mice had a reduced lifespan compared with untreated Msh2(WT) mice (median survival of 127 and 107 days of age, respectively) and developed lymphomas with MSI. CONCLUSION: Azathioprine-induced carcinogenesis in mice depends on the number of functional copies of the Msh2 gene.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Azathioprine/toxicity , Carcinogens/toxicity , DNA Mismatch Repair/genetics , Immunosuppressive Agents/toxicity , Lymphoma/chemically induced , Lymphoma/genetics , MutS Homolog 2 Protein/genetics , Administration, Oral , Animals , DNA, Neoplasm/genetics , Disease Models, Animal , Genotype , Immunohistochemistry , Kaplan-Meier Estimate , Loss of Heterozygosity , Lymphoma/pathology , Mice , Microsatellite Instability/drug effects , Polymerase Chain Reaction , Research Design
9.
Diagn Mol Pathol ; 17(2): 90-3, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18382363

ABSTRACT

The prognosis of patients with colorectal cancer is largely determined by the tumor stage. In this respect, colorectal cancer with lymph node metastases has the worst prognosis. Accordingly, there is considerable clinical interest in understanding the genetic mechanisms underlying metastasis formation. The short arm of chromosome 8 is often lost in colorectal cancer and has been associated with the advanced stages. A common region of deletion has been identified in 8p21, and we investigate here the localization of the putative tumor suppressor gene. A series of 683 sporadic microsatellite stability colorectal tumor samples has been genotyped on 12 microsatellite loci encompassing the common deleted region. Allelic losses were identified in 50% of the cases and 10 break points have been evidenced between D8S1734 and D8S1810, reducing the region of interest to D8S1771-D8S131. Among the 21 genes mapped in this interval, 14 candidate genes have been retained for the sequencing analysis of 48 tumors with 8p allelic loss. No mutation was found, suggesting more complex mechanisms of inactivation or side effects of chromosome arm 8q duplication, which might be up-regulating oncogenes not located within the deleted region.


Subject(s)
Adenocarcinoma/genetics , Chromosomal Instability , Chromosome Breakage , Chromosomes, Human, Pair 8/genetics , Colonic Neoplasms/genetics , Adenocarcinoma/pathology , Chromosome Fragile Sites , Chromosome Mapping , Colonic Neoplasms/pathology , DNA Mutational Analysis , DNA, Neoplasm/analysis , Genes, Tumor Suppressor , Genotype , Humans , Loss of Heterozygosity , Microsatellite Repeats/genetics
10.
Nat Genet ; 39(8): 989-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17618283

ABSTRACT

Using a multistage genetic association approach comprising 7,480 affected individuals and 7,779 controls, we identified markers in chromosomal region 8q24 associated with colorectal cancer. In stage 1, we genotyped 99,632 SNPs in 1,257 affected individuals and 1,336 controls from Ontario. In stages 2-4, we performed serial replication studies using 4,024 affected individuals and 4,042 controls from Seattle, Newfoundland and Scotland. We identified one locus on chromosome 8q24 and another on 9p24 having combined odds ratios (OR) for stages 1-4 of 1.18 (trend; P = 1.41 x 10(-8)) and 1.14 (trend; P = 1.32 x 10(-5)), respectively. Additional analyses in 2,199 affected individuals and 2,401 controls from France and Europe supported the association at the 8q24 locus (OR = 1.16, trend; 95% confidence interval (c.i.): 1.07-1.26; P = 5.05 x 10(-4)). A summary across all seven studies at the 8q24 locus was highly significant (OR = 1.17, c.i.: 1.12-1.23; P = 3.16 x 10(-11)). This locus has also been implicated in prostate cancer.


Subject(s)
Chromosomes, Human, Pair 8 , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Case-Control Studies , Chromosome Mapping , Humans , Linkage Disequilibrium , Middle Aged
11.
Oncogene ; 21(5): 769-77, 2002 Jan 24.
Article in English | MEDLINE | ID: mdl-11850805

ABSTRACT

In this work, the full-length hTERT gene was isolated and the sequence of the previously unknown region in intron 6 as well as that of upstream and downstream hTERT regions was determined. We have shown that intron 6 includes a variable number of tandem repeats (VNTR) of a 38 bp sequence, (hTERT-VNTR 6-1). Eight alleles of hTERT-VNTR 6-1 were identified among 103 unrelated individuals, ranging from 27 to 47 repeats. hTERT-VNTR 2-2 is another new 61 bp minisatellite repeat found in intron 2 of hTERT. At least four alleles of hTERT-VNTR 2-2 can be distinguished. Previous studies have described polymorphisms for minisatellites hTERT-VNTR 2-1, a 42 bp repeat in intron 2, and hTERT-VNTR 6-2, a 36 bp repeat in intron 6. These, together with another minisatellite found in intron 12, add up to five such structures within the hTERT gene. The segregation of hTERT minisatellites was analysed in families, revealing that the VNTRs are transmitted through meiosis following a Mendelian inheritance. Minisatellites in hTERT were also analysed in matching normal and cancer tissues from patients with tumors; in one patient with a kidney tumor, the two VNTRs in intron 6 had undergone concomitant rearrangements. This observation suggests that chromosomal rearrangements implicating these VNTRs may be associated with the activation of telomerase expression in cancer cells.


Subject(s)
Minisatellite Repeats , Polymorphism, Genetic , Telomerase/genetics , Base Sequence , DNA-Binding Proteins , Genes , Genomic Library , Humans , Introns , Molecular Sequence Data , Neoplasms/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...