Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Genom ; 4(7): 100588, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38917803

ABSTRACT

Alterations in the structure and location of telomeres are pivotal in cancer genome evolution. Here, we applied both long-read and short-read genome sequencing to assess telomere repeat-containing structures in cancers and cancer cell lines. Using long-read genome sequences that span telomeric repeats, we defined four types of telomere repeat variations in cancer cells: neotelomeres where telomere addition heals chromosome breaks, chromosomal arm fusions spanning telomere repeats, fusions of neotelomeres, and peri-centromeric fusions with adjoined telomere and centromere repeats. These results provide a framework for the systematic study of telomeric repeats in cancer genomes, which could serve as a model for understanding the somatic evolution of other repetitive genomic elements.


Subject(s)
Neoplasms , Telomere , Humans , Telomere/genetics , Neoplasms/genetics , Cell Line, Tumor , Genome, Human/genetics , Repetitive Sequences, Nucleic Acid/genetics , Centromere/genetics
2.
bioRxiv ; 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38077026

ABSTRACT

Alterations in the structure and location of telomeres are key events in cancer genome evolution. However, previous genomic approaches, unable to span long telomeric repeat arrays, could not characterize the nature of these alterations. Here, we applied both long-read and short-read genome sequencing to assess telomere repeat-containing structures in cancers and cancer cell lines. Using long-read genome sequences that span telomeric repeat arrays, we defined four types of telomere repeat variations in cancer cells: neotelomeres where telomere addition heals chromosome breaks, chromosomal arm fusions spanning telomere repeats, fusions of neotelomeres, and peri-centromeric fusions with adjoined telomere and centromere repeats. Analysis of lung adenocarcinoma genome sequences identified somatic neotelomere and telomere-spanning fusion alterations. These results provide a framework for systematic study of telomeric repeat arrays in cancer genomes, that could serve as a model for understanding the somatic evolution of other repetitive genomic elements.

3.
Nat Commun ; 12(1): 5855, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615869

ABSTRACT

Karyotype alterations have emerged as on-target complications from CRISPR-Cas9 genome editing. However, the events that lead to these karyotypic changes in embryos after Cas9-treatment remain unknown. Here, using imaging and single-cell genome sequencing of 8-cell stage embryos, we track both spontaneous and Cas9-induced karyotype aberrations through the first three divisions of embryonic development. We observe the generation of abnormal structures of the nucleus that arise as a consequence of errors in mitosis, including micronuclei and chromosome bridges, and determine their contribution to common karyotype aberrations including whole chromosome loss that has been recently reported after editing in embryos. Together, these data demonstrate that Cas9-mediated germline genome editing can lead to unwanted on-target side effects, including major chromosome structural alterations that can be propagated over several divisions of embryonic development.


Subject(s)
CRISPR-Cas Systems , Chromosome Structures , Gene Editing/methods , Genomic Instability , Animals , Chromosome Segregation , Embryo, Mammalian , Embryonic Development/genetics , Karyotype , Mice , Whole Genome Sequencing
4.
Nat Genet ; 53(6): 895-905, 2021 06.
Article in English | MEDLINE | ID: mdl-33846636

ABSTRACT

Genome editing has therapeutic potential for treating genetic diseases and cancer. However, the currently most practicable approaches rely on the generation of DNA double-strand breaks (DSBs), which can give rise to a poorly characterized spectrum of chromosome structural abnormalities. Here, using model cells and single-cell whole-genome sequencing, as well as by editing at a clinically relevant locus in clinically relevant cells, we show that CRISPR-Cas9 editing generates structural defects of the nucleus, micronuclei and chromosome bridges, which initiate a mutational process called chromothripsis. Chromothripsis is extensive chromosome rearrangement restricted to one or a few chromosomes that can cause human congenital disease and cancer. These results demonstrate that chromothripsis is a previously unappreciated on-target consequence of CRISPR-Cas9-generated DSBs. As genome editing is implemented in the clinic, the potential for extensive chromosomal rearrangements should be considered and monitored.


Subject(s)
CRISPR-Cas Systems/genetics , Chromothripsis , Gene Editing , Anemia, Sickle Cell/genetics , Antigens, CD34/metabolism , CRISPR-Associated Protein 9/metabolism , Cell Division , Chromosomes, Human/genetics , DNA Cleavage , Genome, Human , Humans , Micronucleus, Germline/genetics , Tumor Suppressor Protein p53/metabolism
5.
Exp Mol Med ; 52(11): 1777-1786, 2020 11.
Article in English | MEDLINE | ID: mdl-33230251

ABSTRACT

Micronuclei are small DNA-containing nuclear structures that are spatially isolated from the main nucleus. They are frequently found in pathologies, including cancer. It was recently shown that these nuclear structures are not only biomarkers of disease but also play an active role in tumor biology. Many consequences of micronucleus formation on tumor biology are dependent on the frequent and irreversible rupture of their nuclear envelopes, which results in the exposure of their DNA contents to the cytoplasm. In this review, we discuss models of defective nuclear envelope deposition on missegregated chromosomes that lead to nuclear envelope rupture. Furthermore, we expound upon the various downstream consequences of micronucleus nuclear envelope rupture on cells. These consequences include a massive DNA rearrangement phenomenon called chromothripsis and activation of the cGAS-STING innate immune signaling pathway, which can be a double-edged sword with tumorigenesis and tumor prevention functions. Although micronuclei are small structures, the impact they have on cells and their microenvironment is quite large.


Subject(s)
Cell Nucleus , Micronuclei, Chromosome-Defective , Nuclear Envelope/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromothripsis , Cytoplasm/metabolism , DNA Damage , Genomic Instability , Humans , Immunity, Innate , Mitosis , Signal Transduction
6.
Annu Rev Genet ; 49: 183-211, 2015.
Article in English | MEDLINE | ID: mdl-26442848

ABSTRACT

Chromosomal rearrangements are generally thought to accumulate gradually over many generations. However, DNA sequencing of cancer and congenital disorders uncovered a new pattern in which multiple rearrangements arise all at once. The most striking example, chromothripsis, is characterized by tens or hundreds of rearrangements confined to a single chromosome or to local regions over a few chromosomes. Genomic analysis of chromothripsis and the search for its biological mechanism have led to new insights on how chromosome segregation errors can generate mutagenesis and changes to the karyotype. Here, we review the genomic features of chromothripsis and summarize recent progress on understanding its mechanism. This includes reviewing new work indicating that one mechanism to generate chromothripsis is through the physical isolation of chromosomes in abnormal nuclear structures (micronuclei). We also discuss connections revealed by recent genomic analysis of cancers between chromothripsis, chromosome bridges, and ring chromosomes.


Subject(s)
Biological Evolution , Chromosomes/genetics , Karyotype , Mutagenesis/physiology , Chromosomes, Human/genetics , Gene Rearrangement , Humans , Micronuclei, Chromosome-Defective , Nuclear Envelope/genetics , Ring Chromosomes
7.
Genes Dev ; 27(23): 2513-30, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298051

ABSTRACT

Recent genome sequencing studies have identified several classes of complex genomic rearrangements that appear to be derived from a single catastrophic event. These discoveries identify ways that genomes can be altered in single large jumps rather than by many incremental steps. Here we compare and contrast these phenomena and examine the evidence that they arise "all at once." We consider the impact of massive chromosomal change for the development of diseases such as cancer and for evolution more generally. Finally, we summarize current models for underlying mechanisms and discuss strategies for testing these models.


Subject(s)
Evolution, Molecular , Gene Rearrangement/genetics , Genome, Human/genetics , Chromosome Aberrations , Genomic Instability , Humans
8.
Genome Res ; 23(5): 762-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23410887

ABSTRACT

Tumor genomes are generally thought to evolve through a gradual accumulation of mutations, but the observation that extraordinarily complex rearrangements can arise through single mutational events suggests that evolution may be accelerated by punctuated changes in genome architecture. To assess the prevalence and origins of complex genomic rearrangements (CGRs), we mapped 6179 somatic structural variation breakpoints in 64 cancer genomes from seven tumor types and screened for clusters of three or more interconnected breakpoints. We find that complex breakpoint clusters are extremely common: 154 clusters comprise 25% of all somatic breakpoints, and 75% of tumors exhibit at least one complex cluster. Based on copy number state profiling, 63% of breakpoint clusters are consistent with being CGRs that arose through a single mutational event. CGRs have diverse architectures including focal breakpoint clusters, large-scale rearrangements joining clusters from one or more chromosomes, and staggeringly complex chromothripsis events. Notably, chromothripsis has a significantly higher incidence in glioblastoma samples (39%) relative to other tumor types (9%). Chromothripsis breakpoints also show significantly elevated intra-tumor allele frequencies relative to simple SVs, which indicates that they arise early during tumorigenesis or confer selective advantage. Finally, assembly and analysis of 4002 somatic and 6982 germline breakpoint sequences reveal that somatic breakpoints show significantly less microhomology and fewer templated insertions than germline breakpoints, and this effect is stronger at CGRs than at simple variants. These results are inconsistent with replication-based models of CGR genesis and strongly argue that nonhomologous repair of concurrently arising DNA double-strand breaks is the predominant mechanism underlying complex cancer genome rearrangements.


Subject(s)
Chromosome Aberrations , Chromosome Breakpoints , Mutation/genetics , Neoplasms/genetics , Base Sequence , DNA Breaks, Double-Stranded , DNA Replication/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/pathology
9.
Cell Stem Cell ; 9(4): 366-73, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21982236

ABSTRACT

The biomedical utility of induced pluripotent stem cells (iPSCs) will be diminished if most iPSC lines harbor deleterious genetic mutations. Recent microarray studies have shown that human iPSCs carry elevated levels of DNA copy number variation compared with those in embryonic stem cells, suggesting that these and other classes of genomic structural variation (SV), including inversions, smaller duplications and deletions, complex rearrangements, and retroelement transpositions, may frequently arise as a consequence of reprogramming. Here we employ whole-genome paired-end DNA sequencing and sensitive mapping algorithms to identify all classes of SV in three fully pluripotent mouse iPSC lines. Despite the improved scope and resolution of this study, we find few spontaneous mutations per line (one or two) and no evidence for endogenous retroelement transposition. These results show that genome stability can persist throughout reprogramming, and argue that it is possible to generate iPSCs lacking gene-disrupting mutations using current reprogramming methods.


Subject(s)
Cellular Reprogramming/genetics , Gene Rearrangement/genetics , Genome/genetics , Genomic Instability/genetics , Induced Pluripotent Stem Cells/metabolism , Retroelements/genetics , Sequence Analysis, DNA/methods , Animals , Base Sequence , Cell Lineage/genetics , Chimera/genetics , DNA Copy Number Variations/genetics , False Negative Reactions , Gene Silencing , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Molecular Sequence Data , Mutagenesis, Insertional/genetics , Organ Specificity/genetics
10.
Genome Res ; 20(5): 623-35, 2010 May.
Article in English | MEDLINE | ID: mdl-20308636

ABSTRACT

Structural variation (SV) is a rich source of genetic diversity in mammals, but due to the challenges associated with mapping SV in complex genomes, basic questions regarding their genomic distribution and mechanistic origins remain unanswered. We have developed an algorithm (HYDRA) to localize SV breakpoints by paired-end mapping, and a general approach for the genome-wide assembly and interpretation of breakpoint sequences. We applied these methods to two inbred mouse strains: C57BL/6J and DBA/2J. We demonstrate that HYDRA accurately maps diverse classes of SV, including those involving repetitive elements such as transposons and segmental duplications; however, our analysis of the C57BL/6J reference strain shows that incomplete reference genome assemblies are a major source of noise. We report 7196 SVs between the two strains, more than two-thirds of which are due to transposon insertions. Of the remainder, 59% are deletions (relative to the reference), 26% are insertions of unlinked DNA, 9% are tandem duplications, and 6% are inversions. To investigate the origins of SV, we characterized 3316 breakpoint sequences at single-nucleotide resolution. We find that approximately 16% of non-transposon SVs have complex breakpoint patterns consistent with template switching during DNA replication or repair, and that this process appears to preferentially generate certain classes of complex variants. Moreover, we find that SVs are significantly enriched in regions of segmental duplication, but that this effect is largely independent of DNA sequence homology and thus cannot be explained by non-allelic homologous recombination (NAHR) alone. This result suggests that the genetic instability of such regions is often the cause rather than the consequence of duplicated genomic architecture.


Subject(s)
Chromosome Breakpoints , Chromosome Mapping , DNA Transposable Elements/genetics , Genetic Variation , Genome , Segmental Duplications, Genomic/genetics , Algorithms , Animals , Base Sequence , Chromosome Mapping/methods , Computational Biology/methods , Female , Genome/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...