Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.129
Filter
1.
Dokl Biochem Biophys ; 517(1): 259-263, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002012

ABSTRACT

Nucleoli form interchromosomal contacts with genes controlling differentiation and carcinogenesis. DUX4 genes specify transcription factor possessing two homeodomains. Previously, using Circular Chromosome Conformation Capture (4С) approach on population of cells, it was demonstrated that DUX4 gene clusters form frequent contacts with nucleoli. It was found also that these contacts are almost completely abolished after heat shock treatment. 4C approach as all ligation-mediated methods is capable to detect rather close interactions between chromatin loops in nuclei. In order to independently confirm the formation and the frequency of the contacts in single cells we used FISH approach. Here, we show that DUX genes in single cells form stable contacts in all tested HEK293T cells. During heat shock, DUX4 genes reversibly move 1-3 µm away from the nuclei. We conclude that interchromosomal contacts formed by nucleoli are strong, dynamic, and reversible, providing both the initiation and maintenance of a differentiated state.


Subject(s)
Cell Nucleolus , Homeodomain Proteins , Humans , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , HEK293 Cells , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , In Situ Hybridization, Fluorescence
2.
J Korean Med Sci ; 39(27): e198, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39015000

ABSTRACT

BACKGROUND: Relatives share more genomic regions than unrelated individuals, with closer relatives sharing more regions. This concept, paired with the increased availability of high-throughput single nucleotide polymorphism (SNP) genotyping technologies, has made it feasible to measure the shared chromosomal regions between individuals to assess their level of relation to each other. However, such techniques have remained in the conceptual rather than practical stages in terms of applying measures or indices. Recently, we developed an index called "genetic distance-based index of chromosomal sharing (GD-ICS)" utilizing large-scale SNP data from Korean family samples and demonstrated its potential for practical applications in kinship determination. In the current study, we present validation results from various real cases demonstrating the utility of this method in resolving complex familial relationships where information obtained from traditional short tandem repeats (STRs) or lineage markers is inconclusive. METHODS: We obtained large-scale SNP data through microarray analysis from Korean individuals involving 13 kinship cases and calculated GD-ICS values using the method described in our previous study. Based on the GD-ICS reference constructed for Korean families, each disputed kinship was evaluated and validated using a combination of traditional STRs and lineage markers. RESULTS: The cases comprised those A) that were found to be inconclusive using the traditional approach, B) for which it was difficult to apply traditional testing methods, and C) that were more conclusively resolved using the GD-ICS method. This method has overcome the limitations faced by traditional STRs in kinship testing, particularly in a paternity case with STR mutational events and in confirming distant kinship where the individual of interest is unavailable for testing. It has also been demonstrated to be effective in identifying various relationships without specific presumptions and in confirming a lack of genetic relatedness between individuals. CONCLUSION: This method has been proven effective in identifying familial relationships across diverse complex and practical scenarios. It is not only useful when traditional testing methods fail to provide conclusive results, but it also enhances the resolution of challenging kinship cases, which suggests its applicability in various types of practical casework.


Subject(s)
Pedigree , Polymorphism, Single Nucleotide , Female , Humans , Male , Chromosomes, Human/genetics , Genotype , Microsatellite Repeats/genetics , Republic of Korea , East Asian People/genetics
3.
Methods Mol Biol ; 2837: 45-58, 2024.
Article in English | MEDLINE | ID: mdl-39044074

ABSTRACT

Hepatitis B virus (HBV) infects hepatocytes that are in the G0/G1 phase with intact nuclear membrane and organized chromosome architecture. In the nucleus of the infected cells, HBV covalently closed circular (ccc) DNA, an episomal minichromosome, serves as the template for all viral transcripts and the reservoir of persistent infection. Nuclear positioning of cccDNA can be assessed by the spatial distance between viral DNA and host chromosomal DNA through Circular Chromosome Conformation Capture (4C) combined with high-throughput sequencing (4C-seq). The 4C-seq analysis relies on proximity ligation and is commonly used for mapping genomic DNA regions that communicate within a host chromosome. The method has been tailored for studying nuclear localization of HBV episomal cccDNA in relation to the host chromosomes. In this study, we present a step-by-step protocol for 4C-seq analysis of HBV infection, including sample collection and fixation, 4C DNA library preparation, sequence library preparation, and data analysis. Although limited by proximity ligation of DNA fragments, 4C-seq analysis provides useful information of HBV localization in 3D genome, and aids the understanding of viral transcription in light of host chromatin conformation.


Subject(s)
DNA, Circular , DNA, Viral , Hepatitis B virus , High-Throughput Nucleotide Sequencing , Hepatitis B virus/genetics , Humans , DNA, Circular/genetics , DNA, Circular/metabolism , DNA, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Hepatitis B/virology , Host-Pathogen Interactions/genetics , Chromosomes/genetics , Gene Library , Chromosomes, Human/genetics , Chromosomes, Human/virology
4.
Sci Adv ; 10(24): eadk4387, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38865460

ABSTRACT

The function of TERRA in the regulation of telomerase in human cells is still debated. While TERRA interacts with telomerase, how it regulates telomerase function remains unknown. Here, we show that TERRA colocalizes with the telomerase RNA subunit hTR in the nucleoplasm and at telomeres during different phases of the cell cycle. We report that TERRA transcripts relocate away from chromosome ends during telomere lengthening, leading to a reduced number of telomeric TERRA-hTR molecules and consequent increase in "TERRA-free" telomerase molecules at telomeres. Using live-cell imaging and super-resolution microscopy, we show that upon transcription, TERRA relocates from its telomere of origin to long chromosome ends. Furthermore, TERRA depletion by antisense oligonucleotides promoted hTR localization to telomeres, leading to increased residence time and extended half-life of hTR molecules at telomeres. Overall, our findings indicate that telomeric TERRA transcripts inhibit telomere elongation by telomerase acting in trans, impairing telomerase access to telomeres that are different from their chromosome end of origin.


Subject(s)
Telomerase , Telomere , Telomerase/metabolism , Telomerase/genetics , Humans , Telomere/metabolism , Telomere/genetics , Telomere Homeostasis , HeLa Cells , RNA/metabolism , RNA/genetics , Transcription, Genetic , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Cell Cycle/genetics , Chromosomes, Human/metabolism , Chromosomes, Human/genetics , DNA-Binding Proteins , Transcription Factors
5.
Cell ; 187(15): 3936-3952.e19, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38936359

ABSTRACT

Duplication is a foundation of molecular evolution and a driver of genomic and complex diseases. Here, we develop a genome editing tool named Amplification Editing (AE) that enables programmable DNA duplication with precision at chromosomal scale. AE can duplicate human genomes ranging from 20 bp to 100 Mb, a size comparable to human chromosomes. AE exhibits activity across various cell types, encompassing diploid, haploid, and primary cells. AE exhibited up to 73.0% efficiency for 1 Mb and 3.4% for 100 Mb duplications, respectively. Whole-genome sequencing and deep sequencing of the junctions of edited sequences confirm the precision of duplication. AE can create chromosomal microduplications within disease-relevant regions in embryonic stem cells, indicating its potential for generating cellular and animal models. AE is a precise and efficient tool for chromosomal engineering and DNA duplication, broadening the landscape of precision genome editing from an individual genetic locus to the chromosomal scale.


Subject(s)
Gene Duplication , Gene Editing , Genome, Human , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , DNA/genetics , Animals , Embryonic Stem Cells/metabolism , Chromosomes, Human/genetics
6.
Methods Mol Biol ; 2825: 137-150, 2024.
Article in English | MEDLINE | ID: mdl-38913307

ABSTRACT

Chromosome banding can be defined as the lengthwise variation in staining properties along a chromosome stained with a dye. Chromosome banding became more practical in the early 1970s and is an essential technique used in karyotyping to identify human chromosomes for both clinical and research purposes. Most importantly, karyotyping is now considered a mandatory investigation of all newly diagnosed leukemias. Some banding methods, such as Giemsa (G)-, reverse (R)-, and centromere (C)-banding, still contribute greatly by being used as a routine procedure in clinical cytogenetic laboratory nowadays. Each chromosome has a unique sequence of bar code-like stripes, allowing the identification of individual homologues and the recognition of structural abnormalities through analyzing the disruption of the normal banding pattern at specific landmarks, regions, and bands as described in the ideogram. Since the quality of metaphases obtained from malignant cells is generally inferior to normal constitutional cells for karyotyping, a practical and accurate chromosome identification training guide is indispensable for a trainee or newly employed cytogenetic technologist in a cancer cytogenetic laboratory. The most common and currently used banding methods and chromosome recognition guide for distinguishable bands of each chromosome are described in detail in this chapter with an aim to facilitate quick and accurate karyotyping in cancer cells.


Subject(s)
Chromosome Banding , Karyotyping , Humans , Karyotyping/methods , Chromosomes, Human/genetics , Metaphase
7.
Methods Mol Biol ; 2825: 213-237, 2024.
Article in English | MEDLINE | ID: mdl-38913312

ABSTRACT

Three-dimensional structured illumination microscopy (3D-SIM) and fluorescence in situ hybridization on three-dimensional preserved cells (3D-FISH) have proven to be robust and efficient methodologies for analyzing nuclear architecture and profiling the genome's topological features. These methods have allowed the simultaneous visualization and evaluation of several target structures at super-resolution. In this chapter, we focus on the application of 3D-SIM for the visualization of 3D-FISH preparations of chromosomes in interphase, known as Chromosome Territories (CTs). We provide a workflow and detailed guidelines for sample preparation, image acquisition, and image analysis to obtain quantitative measurements for profiling chromosome topological features. In parallel, we address a practical example of these protocols in the profiling of CTs 9 and 22 involved in the translocation t(9;22) in Chronic Myeloid Leukemia (CML). The profiling of chromosome topological features described in this chapter allowed us to characterize a large-scale topological disruption of CTs 9 and 22 that correlates directly with patients' response to treatment and as a possible potential change in the inheritance systems. These findings open new insights into how the genome structure is associated with the response to cancer treatments, highlighting the importance of microscopy in analyzing the topological features of the genome.


Subject(s)
Imaging, Three-Dimensional , In Situ Hybridization, Fluorescence , Humans , In Situ Hybridization, Fluorescence/methods , Imaging, Three-Dimensional/methods , Translocation, Genetic , Chromosomes/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Interphase/genetics , Chromosomes, Human/genetics , Image Processing, Computer-Assisted/methods
8.
Nat Commun ; 15(1): 3905, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724522

ABSTRACT

Glioblastoma multiforme (GBM) encompasses brain malignancies marked by phenotypic and transcriptional heterogeneity thought to render these tumors aggressive, resistant to therapy, and inevitably recurrent. However, little is known about how the spatial organization of GBM genomes underlies this heterogeneity and its effects. Here, we compile a cohort of 28 patient-derived glioblastoma stem cell-like lines (GSCs) known to reflect the properties of their tumor-of-origin; six of these were primary-relapse tumor pairs from the same patient. We generate and analyze 5 kbp-resolution chromosome conformation capture (Hi-C) data from all GSCs to systematically map thousands of standalone and complex structural variants (SVs) and the multitude of neoloops arising as a result. By combining Hi-C, histone modification, and gene expression data with chromatin folding simulations, we explain how the pervasive, uneven, and idiosyncratic occurrence of neoloops sustains tumor-specific transcriptional programs via the formation of new enhancer-promoter contacts. We also show how even moderately recurrent neoloops can relate to patient-specific vulnerabilities. Together, our data provide a resource for dissecting GBM biology and heterogeneity, as well as for informing therapeutic approaches.


Subject(s)
Brain Neoplasms , Chromatin , Gene Expression Regulation, Neoplastic , Glioblastoma , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Chromatin/metabolism , Chromatin/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Genetic Heterogeneity , Promoter Regions, Genetic/genetics , Transcription, Genetic , Enhancer Elements, Genetic/genetics , Chromosomes, Human/genetics
9.
Comput Biol Med ; 177: 108601, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776728

ABSTRACT

Automated karyotyping is of great importance for cytogenetic research, as it speeds up the process for cytogeneticists through incorporating AI-driven automated segmentation and classification techniques. Existing frameworks confront two primary issues: Firstly the necessity for instance-level data annotation with either detection bounding boxes or semantic masks for training, and secondly, its poor robustness particularly when confronted with domain shifts. In this work, we first propose an accurate segmentation framework, namely KaryoXpert. This framework leverages the strengths of both morphology algorithms and deep learning models, allowing for efficient training that breaks the limit for the acquirement of manually labeled ground-truth mask annotations. Additionally, we present an accurate classification model based on metric learning, designed to overcome the challenges posed by inter-class similarity and batch effects. Our framework exhibits state-of-the-art performance with exceptional robustness in both chromosome segmentation and classification. The proposed KaryoXpert framework showcases its capacity for instance-level chromosome segmentation even in the absence of annotated data, offering novel insights into the research for automated chromosome segmentation. The proposed method has been successfully deployed to support clinical karyotype diagnosis.


Subject(s)
Karyotyping , Humans , Karyotyping/methods , Metaphase , Algorithms , Chromosomes, Human/genetics , Image Processing, Computer-Assisted/methods , Deep Learning
10.
Science ; 384(6695): 533-539, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38603523

ABSTRACT

Short telomeres cause age-related disease, and long telomeres contribute to cancer; however, the mechanisms regulating telomere length are unclear. We developed a nanopore-based method, which we call Telomere Profiling, to determine telomere length at nearly single-nucleotide resolution. Mapping telomere reads to chromosome ends showed chromosome end-specific length distributions that could differ by more than six kilobases. Examination of telomere lengths in 147 individuals revealed that certain chromosome ends were consistently longer or shorter. The same rank order was found in newborn cord blood, suggesting that telomere length is determined at birth and that chromosome end-specific telomere length differences are maintained as telomeres shorten with age. Telomere Profiling makes precision investigation of telomere length widely accessible for laboratory, clinical, and drug discovery efforts and will allow deeper insights into telomere biology.


Subject(s)
Chromosome Mapping , Nanopore Sequencing , Telomere Homeostasis , Telomere Shortening , Telomere , Humans , Male , Chromosomes, Human/genetics , Fetal Blood , Nanopore Sequencing/methods , Telomere/genetics , Telomere Homeostasis/genetics , Telomere Shortening/genetics , Chromosome Mapping/methods
11.
Int J Radiat Biol ; 100(5): 709-714, 2024.
Article in English | MEDLINE | ID: mdl-38394348

ABSTRACT

PURPOSE: Networking with other biodosimetry laboratories is necessary to assess the radiation exposure of many individuals in large-scale radiological accidents. The Korea biodosimetry network, K-BioDos, prepared harmonized scoring guidelines for dicentric chromosome assay to obtain homogeneous results within the network and investigated the efficiency of the guidelines. MATERIALS AND METHODS: Three laboratories in K-BioDos harmonized the scoring guidelines for dicentric chromosome assay. The results of scoring dicentric chromosomes using the harmonized scoring guidelines were compared with the laboratories' results using their own methods. Feedback was collected from the scorers following the three intercomparison exercises in 3 consecutive years. RESULTS: K-BioDos members showed comparable capacity to score dicentrics in the three exercises. However, the results of the K-BioDos guidelines showed no significant improvement over those of the scorers' own methods. According to the scorers, our harmonized guidelines led to more rejected metaphases and ultimately decreased the number of scorable metaphases compared with their own methods. Moreover, the scoring time was sometimes longer with the K-BioDos protocol because some scorers were not yet familiar with the guidelines, though most scorers reported that the time decreased or was unchanged. These challenges may cause low adherence to the guidelines. Most scorers expressed willingness to use the guidelines to select scorable metaphases or identify dicentrics for other biodosimetry works, whereas one did not want to use it due to the difference from their calibration curves. CONCLUSIONS: We identified potential resistance to following the harmonized guidelines and received requests for more detailed methods. Our findings suggest that the harmonized criteria should be continually updated, and education and training should be provided for all scorers. These changes could allow members within the biodosimetry network to successfully collaborate and support each other in large-scale radiological accidents.


Subject(s)
Chromosome Aberrations , Republic of Korea , Humans , Chromosomes, Human/genetics , Chromosomes, Human/radiation effects
12.
PLoS Genet ; 19(8): e1010399, 2023 08.
Article in English | MEDLINE | ID: mdl-37578977

ABSTRACT

Evidence of interbreeding between archaic hominins and humans comes from methods that infer the locations of segments of archaic haplotypes, or 'archaic coverage' using the genomes of people living today. As more estimates of archaic coverage have emerged, it has become clear that most of this coverage is found on the autosomes- very little is retained on chromosome X. Here, we summarize published estimates of archaic coverage on autosomes and chromosome X from extant human samples. We find on average 7 times more archaic coverage on autosomes than chromosome X, and identify broad continental patterns in this ratio: greatest in European samples, and least in South Asian samples. We also perform extensive simulation studies to investigate how the amount of archaic coverage, lengths of coverage, and rates of purging of archaic coverage are affected by sex-bias caused by an unequal sex ratio within the archaic introgressors. Our results generally confirm that, with increasing male sex-bias, less archaic coverage is retained on chromosome X. Ours is the first study to explicitly model such sex-bias and its potential role in creating the dearth of archaic coverage on chromosome X.


Subject(s)
Genetic Introgression , Genome, Human , Hominidae , X Chromosome , Animals , Humans , Male , Asian People/genetics , Genome , Genome, Human/genetics , Hominidae/genetics , Neanderthals/genetics , X Chromosome/genetics , Sex Factors , Haplotypes/genetics , Genetic Introgression/genetics , Chromosomes, Human/genetics , Female , South Asian People/genetics , European People/genetics
13.
Nature ; 620(7972): 209-217, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37438531

ABSTRACT

The human genome functions as a three-dimensional chromatin polymer, driven by a complex collection of chromosome interactions1-3. Although the molecular rules governing these interactions are being quickly elucidated, relatively few proteins regulating this process have been identified. Here, to address this gap, we developed high-throughput DNA or RNA labelling with optimized Oligopaints (HiDRO)-an automated imaging pipeline that enables the quantitative measurement of chromatin interactions in single cells across thousands of samples. By screening the human druggable genome, we identified more than 300 factors that influence genome folding during interphase. Among these, 43 genes were validated as either increasing or decreasing interactions between topologically associating domains. Our findings show that genetic or chemical inhibition of the ubiquitous kinase GSK3A leads to increased long-range chromatin looping interactions in a genome-wide and cohesin-dependent manner. These results demonstrate the importance of GSK3A signalling in nuclear architecture and the use of HiDRO for identifying mechanisms of spatial genome organization.


Subject(s)
Chromatin , Chromosome Positioning , Chromosomes, Human , Genome, Human , Glycogen Synthase Kinases , High-Throughput Screening Assays , Single-Cell Analysis , Humans , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , Chromosome Positioning/drug effects , Chromosomes, Human/drug effects , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/analysis , DNA/metabolism , Genome, Human/drug effects , Genome, Human/genetics , Glycogen Synthase Kinases/antagonists & inhibitors , Glycogen Synthase Kinases/deficiency , Glycogen Synthase Kinases/genetics , High-Throughput Screening Assays/methods , Interphase , Reproducibility of Results , RNA/analysis , RNA/metabolism , Signal Transduction/drug effects , Single-Cell Analysis/methods , Cohesins
14.
Nature ; 618(7967): 1041-1048, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37165191

ABSTRACT

Complex genome rearrangements can be generated by the catastrophic pulverization of missegregated chromosomes trapped within micronuclei through a process known as chromothripsis1-5. As each chromosome contains a single centromere, it remains unclear how acentric fragments derived from shattered chromosomes are inherited between daughter cells during mitosis6. Here we tracked micronucleated chromosomes with live-cell imaging and show that acentric fragments cluster in close spatial proximity throughout mitosis for asymmetric inheritance by a single daughter cell. Mechanistically, the CIP2A-TOPBP1 complex prematurely associates with DNA lesions within ruptured micronuclei during interphase, which poises pulverized chromosomes for clustering upon mitotic entry. Inactivation of CIP2A-TOPBP1 caused acentric fragments to disperse throughout the mitotic cytoplasm, stochastically partition into the nucleus of both daughter cells and aberrantly misaccumulate as cytoplasmic DNA. Mitotic clustering facilitates the reassembly of acentric fragments into rearranged chromosomes lacking the extensive DNA copy-number losses that are characteristic of canonical chromothripsis. Comprehensive analysis of pan-cancer genomes revealed clusters of DNA copy-number-neutral rearrangements-termed balanced chromothripsis-across diverse tumour types resulting in the acquisition of known cancer driver events. Thus, distinct patterns of chromothripsis can be explained by the spatial clustering of pulverized chromosomes from micronuclei.


Subject(s)
Chromosomes, Human , Chromothripsis , Micronuclei, Chromosome-Defective , Mitosis , Humans , Centromere , Chromosomes, Human/genetics , DNA/genetics , DNA/metabolism , DNA Copy Number Variations , Interphase , Mitosis/genetics , Neoplasms/genetics
15.
Nature ; 617(7960): 335-343, 2023 05.
Article in English | MEDLINE | ID: mdl-37165241

ABSTRACT

The short arms of the human acrocentric chromosomes 13, 14, 15, 21 and 22 (SAACs) share large homologous regions, including ribosomal DNA repeats and extended segmental duplications1,2. Although the resolution of these regions in the first complete assembly of a human genome-the Telomere-to-Telomere Consortium's CHM13 assembly (T2T-CHM13)-provided a model of their homology3, it remained unclear whether these patterns were ancestral or maintained by ongoing recombination exchange. Here we show that acrocentric chromosomes contain pseudo-homologous regions (PHRs) indicative of recombination between non-homologous sequences. Utilizing an all-to-all comparison of the human pangenome from the Human Pangenome Reference Consortium4 (HPRC), we find that contigs from all of the SAACs form a community. A variation graph5 constructed from centromere-spanning acrocentric contigs indicates the presence of regions in which most contigs appear nearly identical between heterologous acrocentric chromosomes in T2T-CHM13. Except on chromosome 15, we observe faster decay of linkage disequilibrium in the pseudo-homologous regions than in the corresponding short and long arms, indicating higher rates of recombination6,7. The pseudo-homologous regions include sequences that have previously been shown to lie at the breakpoint of Robertsonian translocations8, and their arrangement is compatible with crossover in inverted duplications on chromosomes 13, 14 and 21. The ubiquity of signals of recombination between heterologous acrocentric chromosomes seen in the HPRC draft pangenome suggests that these shared sequences form the basis for recurrent Robertsonian translocations, providing sequence and population-based confirmation of hypotheses first developed from cytogenetic studies 50 years ago9.


Subject(s)
Centromere , Chromosomes, Human , Recombination, Genetic , Humans , Centromere/genetics , Chromosomes, Human/genetics , DNA, Ribosomal/genetics , Recombination, Genetic/genetics , Translocation, Genetic/genetics , Cytogenetics , Telomere/genetics
16.
Nucleic Acids Res ; 51(9): 4237-4251, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36864748

ABSTRACT

Human papillomavirus (HPV) integration is a critical step in cervical cancer development; however, the oncogenic mechanism at the genome-wide transcriptional level is still poorly understood. In this study, we employed integrative analysis on multi-omics data of six HPV-positive and three HPV-negative cell lines. Through HPV integration detection, super-enhancer (SE) identification, SE-associated gene expression and extrachromosomal DNA (ecDNA) investigation, we aimed to explore the genome-wide transcriptional influence of HPV integration. We identified seven high-ranking cellular SEs generated by HPV integration in total (the HPV breakpoint-induced cellular SEs, BP-cSEs), leading to intra-chromosomal and inter-chromosomal regulation of chromosomal genes. The pathway analysis revealed that the dysregulated chromosomal genes were correlated to cancer-related pathways. Importantly, we demonstrated that BP-cSEs existed in the HPV-human hybrid ecDNAs, explaining the above transcriptional alterations. Our results suggest that HPV integration generates cellular SEs that function as ecDNA to regulate unconstrained transcription, expanding the tumorigenic mechanism of HPV integration and providing insights for developing new diagnostic and therapeutic strategies.


Subject(s)
DNA , Enhancer Elements, Genetic , Genome, Human , Human Papillomavirus Viruses , Papillomavirus Infections , Transcription, Genetic , Uterine Cervical Neoplasms , Virus Integration , Female , Humans , Human Papillomavirus Viruses/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Virus Integration/genetics , Enhancer Elements, Genetic/genetics , DNA/genetics , DNA/metabolism , Genome, Human/genetics , Carcinogenesis , Chromosome Breakpoints , Chromosomes, Human/genetics
17.
Nature ; 615(7954): 925-933, 2023 03.
Article in English | MEDLINE | ID: mdl-36922594

ABSTRACT

Whole-genome doubling (WGD) is a recurrent event in human cancers and it promotes chromosomal instability and acquisition of aneuploidies1-8. However, the three-dimensional organization of chromatin in WGD cells and its contribution to oncogenic phenotypes are currently unknown. Here we show that in p53-deficient cells, WGD induces loss of chromatin segregation (LCS). This event is characterized by reduced segregation between short and long chromosomes, A and B subcompartments and adjacent chromatin domains. LCS is driven by the downregulation of CTCF and H3K9me3 in cells that bypassed activation of the tetraploid checkpoint. Longitudinal analyses revealed that LCS primes genomic regions for subcompartment repositioning in WGD cells. This results in chromatin and epigenetic changes associated with oncogene activation in tumours ensuing from WGD cells. Notably, subcompartment repositioning events were largely independent of chromosomal alterations, which indicates that these were complementary mechanisms contributing to tumour development and progression. Overall, LCS initiates chromatin conformation changes that ultimately result in oncogenic epigenetic and transcriptional modifications, which suggests that chromatin evolution is a hallmark of WGD-driven cancer.


Subject(s)
Chromatin , Chromosome Aberrations , Chromosome Segregation , Chromosomes, Human , Genome, Human , Neoplasms , Humans , Chromatin/genetics , Chromatin/metabolism , Neoplasms/genetics , Chromosomes, Human/genetics , Genome, Human/genetics , Chromosome Segregation/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Disease Progression , Transcription, Genetic , Gene Expression Regulation, Neoplastic
18.
Annu Rev Genomics Hum Genet ; 24: 63-83, 2023 08 25.
Article in English | MEDLINE | ID: mdl-36854315

ABSTRACT

The p-arms of the five human acrocentric chromosomes bear nucleolar organizer regions (NORs) comprising ribosomal gene (rDNA) repeats that are organized in a homogeneous tandem array and transcribed in a telomere-to-centromere direction. Precursor ribosomal RNA transcripts are processed and assembled into ribosomal subunits, the nucleolus being the physical manifestation of this process. I review current understanding of nucleolar chromosome biology and describe current exploration into a role for the NOR chromosomal context. Full DNA sequences for acrocentric p-arms are now emerging, aided by the current revolution in long-read sequencing and genome assembly. Acrocentric p-arms vary from 10.1 to 16.7 Mb, accounting for ∼2.2% of the genome. Bordering rDNA arrays, distal junctions, and proximal junctions are shared among the p-arms, with distal junctions showing evidence of functionality. The remaining p-arm sequences comprise multiple satellite DNA classes and segmental duplications that facilitate recombination between heterologous chromosomes, which is likely also involved in Robertsonian translocations.


Subject(s)
Chromosomes, Human , Nucleolus Organizer Region , Humans , Chromosomes, Human/genetics , Chromosomes , Cell Nucleolus/genetics , Centromere , DNA, Ribosomal/genetics
19.
Nature ; 612(7940): 564-572, 2022 12.
Article in English | MEDLINE | ID: mdl-36477537

ABSTRACT

Higher-order chromatin structure is important for the regulation of genes by distal regulatory sequences1,2. Structural variants (SVs) that alter three-dimensional (3D) genome organization can lead to enhancer-promoter rewiring and human disease, particularly in the context of cancer3. However, only a small minority of SVs are associated with altered gene expression4,5, and it remains unclear why certain SVs lead to changes in distal gene expression and others do not. To address these questions, we used a combination of genomic profiling and genome engineering to identify sites of recurrent changes in 3D genome structure in cancer and determine the effects of specific rearrangements on oncogene activation. By analysing Hi-C data from 92 cancer cell lines and patient samples, we identified loci affected by recurrent alterations to 3D genome structure, including oncogenes such as MYC, TERT and CCND1. By using CRISPR-Cas9 genome engineering to generate de novo SVs, we show that oncogene activity can be predicted by using 'activity-by-contact' models that consider partner region chromatin contacts and enhancer activity. However, activity-by-contact models are only predictive of specific subsets of genes in the genome, suggesting that different classes of genes engage in distinct modes of regulation by distal regulatory elements. These results indicate that SVs that alter 3D genome organization are widespread in cancer genomes and begin to illustrate predictive rules for the consequences of SVs on oncogene activation.


Subject(s)
Genomic Structural Variation , Neoplasms , Oncogene Proteins , Oncogenes , Humans , Chromatin/genetics , Gene Rearrangement/genetics , Genomic Structural Variation/genetics , Neoplasms/genetics , Neoplasms/pathology , Oncogenes/genetics , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Chromosomes, Human/genetics , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , Models, Genetic
20.
Sci Rep ; 12(1): 22097, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543843

ABSTRACT

The dicentric chromosome assay is the "gold standard" in biodosimetry for estimating radiation exposure. However, its large-scale deployment is limited owing to its time-consuming nature and requirement for expert reviewers. Therefore, a recently developed automated system was evaluated for the dicentric chromosome assay. A previously constructed deep learning-based automatic dose-estimation system (DLADES) was used to construct dose curves and calculate estimated doses. Blood samples from two donors were exposed to cobalt-60 gamma rays (0-4 Gy, 0.8 Gy/min). The DLADES efficiently identified monocentric and dicentric chromosomes but showed impaired recognition of complete cells with 46 chromosomes. We estimated the chromosome number of each "Accepted" sample in the DLADES and sorted similar-quality images by removing outliers using the 1.5IQR method. Eleven of the 12 data points followed Poisson distribution. Blind samples were prepared for each dose to verify the accuracy of the estimated dose generated by the curve. The estimated dose was calculated using Merkle's method. The actual dose for each sample was within the 95% confidence limits of the estimated dose. Sorting similar-quality images using chromosome numbers is crucial for the automated dicentric chromosome assay. We successfully constructed a dose-response curve and determined the estimated dose using the DLADES.


Subject(s)
Deep Learning , Radiometry , Humans , Radiometry/methods , Chromosome Aberrations , Gamma Rays , Chromosomes, Human/genetics , Dose-Response Relationship, Radiation
SELECTION OF CITATIONS
SEARCH DETAIL