1
|
Lee RRQ, Chae E. Monkeys at Rigged Typewriters: A Population and Network View of Plant Immune System Incompatibility. ANNUAL REVIEW OF PLANT BIOLOGY 2025; 76:523-550. [PMID: 40030162 DOI: 10.1146/annurev-arplant-083023-041225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Immune system incompatibilities between naturally occurring genomic variants underlie many hybrid defects in plants and present a barrier for crop improvement. In this review, we approach immune system incompatibilities from pan-genomic and network perspectives. Pan-genomes offer insights into how natural variation shapes the evolutionary landscape of immune system incompatibilities, and through it, selection, polymorphisms, and recombination resistance emerge as common features that synergistically drive these incompatibilities. By contextualizing incompatibilities within the immune network, immune receptor promiscuity, complex dysregulation, and single-point failure appear to be recurrent themes of immune system defects. As geneticists break genes to investigate their function, so can we investigate broken immune systems to enrich our understanding of plant immune systems and work toward improving them.
Collapse
Affiliation(s)
- Rachelle R Q Lee
- Department of Biological Sciences, National University of Singapore, Singapore;
| | - Eunyoung Chae
- Department of Biological Sciences, National University of Singapore, Singapore;
- Department of Biology, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
2
|
Raeisi Dehkordi S, Wong ITL, Ni J, Luebeck J, Zhu K, Prasad G, Krockenberger L, Xu G, Chowdhury B, Rajkumar U, Caplin A, Muliaditan D, Gnanasekar A, Coruh C, Jin Q, Turner K, Teo SX, Pang AWC, Alexandrov LB, Chua CEL, Furnari FB, Maciejowski J, Paulson TG, Law JA, Chang HY, Yue F, DasGupta R, Zhao J, Mischel PS, Bafna V. Breakage fusion bridge cycles drive high oncogene number with moderate intratumoural heterogeneity. Nat Commun 2025; 16:1497. [PMID: 39929823 PMCID: PMC11811125 DOI: 10.1038/s41467-025-56670-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
Oncogene amplification is a key driver of cancer pathogenesis. Both breakage fusion bridge (BFB) cycles and extrachromosomal DNA (ecDNA) can lead to high oncogene copy numbers, but the impact of BFB amplifications on intratumoral heterogeneity, treatment response, and patient survival remains poorly understood due to detection challenges with DNA sequencing. We introduce an algorithm, OM2BFB, designed to detect and reconstruct BFB amplifications using optical genome mapping (OGM). OM2BFB demonstrates high precision (>93%) and recall (92%) in identifying BFB amplifications across cancer cell lines, patient-derived xenograft models, and primary tumors. Comparisons using OGM reveal that BFB detection with our AmpliconSuite toolkit for short-read sequencing also achieves high precision, though with reduced sensitivity. We identify 371 BFB events through whole genome sequencing of 2557 primary tumors and cancer cell lines. BFB amplifications are prevalent in cervical, head and neck, lung, and esophageal cancers, but rare in brain cancers. Genes amplified through BFB exhibit lower expression variance, with limited potential for regulatory adaptation compared to ecDNA-amplified genes. Tumors with BFB amplifications (BFB(+)) show reduced structural heterogeneity in amplicons and delayed resistance onset relative to ecDNA(+) tumors. These findings highlight ecDNA and BFB amplifications as distinct oncogene amplification mechanisms with differing biological characteristics, suggesting distinct avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Siavash Raeisi Dehkordi
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Ivy Tsz-Lo Wong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jing Ni
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Gino Prasad
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Lena Krockenberger
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Guanghui Xu
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Biswanath Chowdhury
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Utkrisht Rajkumar
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Ann Caplin
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Daniel Muliaditan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Aditi Gnanasekar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Ceyda Coruh
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- ClearNote Health, San Diego, CA, 92121, USA
| | - Qiushi Jin
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | | | - Shu Xian Teo
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore, 138672, Republic of Singapore
| | | | - Ludmil B Alexandrov
- Moores Cancer Center, UC San Diego Health, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California at San Diego, La Jolla, CA, USA
| | - Christelle En Lin Chua
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore, 138672, Republic of Singapore
| | - Frank B Furnari
- Department of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Maciejowski
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas G Paulson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julie A Law
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Ramanuj DasGupta
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
- School of Cancer Sciences, University of Glasgow; Senior Group Leader, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Jean Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Paul S Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA.
- Halıcıoğlu Data Science Institute, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Liu G, Liu Q, Zhao J, Luo R, Wan Y, Luo Z. Integrated Analysis of Single-Cell and Bulk RNA Sequencing Reveals HSD3B7 as a Prognostic Biomarker and Potential Therapeutic Target in ccRCC. Int J Mol Sci 2024; 25:12929. [PMID: 39684640 DOI: 10.3390/ijms252312929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common kidney malignancy, with a poor prognosis for advanced-stage patients. Identifying key biomarkers involved in tumor progression is crucial for improving treatment outcomes. In this study, we employed an integrated approach combining single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) to identify biomarkers associated with ccRCC progression and prognosis. Single-cell transcriptomic data were obtained from publicly available datasets, and genes related to tumor progression were screened using Monocle2. Bulk RNA-seq data for ccRCC were retrieved from The Cancer Genome Atlas (TCGA) and integrated with scRNA-seq data to explore tumor heterogeneity. We identified 3 beta-hydroxy steroid dehydrogenase type 7 (HSD3B7) as a candidate biomarker for ccRCC, associated with poor overall survival, disease-specific survival, and progression-free interval. Elevated HSD3B7 expression correlated with aggressive clinical features such as advanced TNM stages, histologic grades, and metastasis. Functional studies demonstrated that HSD3B7 promotes cell proliferation, migration, and invasion in vitro, while its silencing significantly inhibits tumor growth in vivo. Our findings reveal that HSD3B7 is a novel biomarker for ccRCC, providing insights into its role in tumor progression and potential as a target for therapy. This study highlights the value of integrating scRNA-seq and bulk RNA-seq data to uncover key regulators of tumor biology and lays the foundation for developing personalized therapeutic strategies for ccRCC patients.
Collapse
Affiliation(s)
- Guicen Liu
- Molecular Medicine and Cancer Research Center, School of Basic Medical Science, Chongqing Medical University, 400016 Chongqing, China
| | - Qichen Liu
- College of Paediatrics, Chongqing Medical University, 400016 Chongqing, China
| | - Jiawei Zhao
- Molecular Medicine and Cancer Research Center, School of Basic Medical Science, Chongqing Medical University, 400016 Chongqing, China
| | - Ruyue Luo
- Molecular Medicine and Cancer Research Center, School of Basic Medical Science, Chongqing Medical University, 400016 Chongqing, China
| | - Yuan Wan
- Molecular Medicine and Cancer Research Center, School of Basic Medical Science, Chongqing Medical University, 400016 Chongqing, China
| | - Zhongli Luo
- Molecular Medicine and Cancer Research Center, School of Basic Medical Science, Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
4
|
Dehkordi SR, Wong ITL, Ni J, Luebeck J, Zhu K, Prasad G, Krockenberger L, Xu G, Chowdhury B, Rajkumar U, Caplin A, Muliaditan D, Coruh C, Jin Q, Turner K, Teo SX, Pang AWC, Alexandrov LB, Chua CEL, Furnari FB, Paulson TG, Law JA, Chang HY, Yue F, DasGupta R, Zhao J, Mischel PS, Bafna V. Breakage fusion bridge cycles drive high oncogene copy number, but not intratumoral genetic heterogeneity or rapid cancer genome change. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571349. [PMID: 38168210 PMCID: PMC10760206 DOI: 10.1101/2023.12.12.571349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Oncogene amplification is a major driver of cancer pathogenesis. Breakage fusion bridge (BFB) cycles, like extrachromosomal DNA (ecDNA), can lead to high copy numbers of oncogenes, but their impact on intratumoral heterogeneity, treatment response, and patient survival are not well understood due to difficulty in detecting them by DNA sequencing. We describe a novel algorithm that detects and reconstructs BFB amplifications using optical genome maps (OGMs), called OM2BFB. OM2BFB showed high precision (>93%) and recall (92%) in detecting BFB amplifications in cancer cell lines, PDX models and primary tumors. OM-based comparisons demonstrated that short-read BFB detection using our AmpliconSuite (AS) toolkit also achieved high precision, albeit with reduced sensitivity. We detected 371 BFB events using whole genome sequences from 2,557 primary tumors and cancer lines. BFB amplifications were preferentially found in cervical, head and neck, lung, and esophageal cancers, but rarely in brain cancers. BFB amplified genes show lower variance of gene expression, with fewer options for regulatory rewiring relative to ecDNA amplified genes. BFB positive (BFB (+)) tumors showed reduced heterogeneity of amplicon structures, and delayed onset of resistance, relative to ecDNA(+) tumors. EcDNA and BFB amplifications represent contrasting mechanisms to increase the copy numbers of oncogene with markedly different characteristics that suggest different routes for intervention.
Collapse
Affiliation(s)
- Siavash Raeisi Dehkordi
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Ivy Tsz-Lo Wong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jing Ni
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, San Diego, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Gino Prasad
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Lena Krockenberger
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Guanghui Xu
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Biswanath Chowdhury
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Utkrisht Rajkumar
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Ann Caplin
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Daniel Muliaditan
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ceyda Coruh
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- ClearNote Health, San Diego, CA 92121 USA
| | - Qiushi Jin
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | | | - Shu Xian Teo
- Singapore Nuclear Research and Safety Initiative, National University of Singapore
| | | | - Ludmil B Alexandrov
- Moores Cancer Center, UC San Diego Health, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California at San Diego, La Jolla, CA, USA
| | | | - Frank B Furnari
- Department of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Thomas G Paulson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julie A Law
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jean Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
| | - Paul S Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California at San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Li Z, Gao Y, Du L, Yuan Y, Huang W, Fu X, Huang Y, Zhang X, You F, Li S. Anti-inflammatory and anti-apoptotic effects of Shaoyao decoction on X-ray radiation-induced enteritis of C57BL/6 mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115158. [PMID: 35245630 DOI: 10.1016/j.jep.2022.115158] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/24/2022] [Accepted: 02/27/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a typical heat-clearing prescription, Shaoyao decoction (SYD) has a robust function of clearing viscera heat for the treatment of several intestinal discomfort symptoms. Clinical evidence indicated that it had the potential to cure radiation enteritis. However, its underlying mechanisms remain unclear. AIM OF THE STUDY The present study was designed to probe the protective effects and the involved mechanisms of SYD on X-ray radiation-induced enteritis of C57BL/6 mice. MATERIALS AND METHODS X-ray irradiation were used to establish the radiation enteritis model. Forty-eight male C57BL/6 mice (20 ± 2 g) were randomly divided into six groups: the control group, model group, dexamethasone group (DEX, 0.12 mg/kg) and SYD groups (0.12, 0.24 and 0.36 g/mL), respectively. All mice (except the control group) were intragastrically administrated for a continuous 7 days. H&E and Masson staining were employed to evaluate the morphological and collagen fibers changes of the colon. ELISA was performed to assess the levels of MDA, SOD, COX, LPS, IL-6, IL-1β and TNF-α in serum. Moreover, TUNEL fluorescence, western blot and qRT-PCR were used to detect the levels of apoptosis-related proteins and genes of Dclk-1, ATM, MRE-11, Bcl-2, Bax, Caspase-3, and Cyto-c. Furthermore, immunofluorescent staining was applied to detect the protein levels of p53 and Claudin-1 in colon. RESULTS Treatment with SYD decreased the exfoliated and necrotic epithelial cells and prevent the proliferate from damaged fibrous tissue in the crypt layer of mucos. The levels of serum peroxidation and pro-inflammatory cytokines (MDA, COX, LPS, IL-6, IL-1β and TNF-α) were obviously inhibited, while SOD sharply increased in serum after administration. Moreover, SYD can significantly ameliorate the apoptosis of colon cells, evidenced by the reduced positive expression of TUNEL staining. Meanwhile, the results of qRT-PCR and western blot demonstrated that SYD can dramatically stimulate the expression of genes and proteins Dclk-1, ATM and MRE-11, thus promoting the expression of mitochondrial pro-apoptotic proteins Bax, Caspase-3 and Cyto-c, while increasing the level of anti-apoptotic protein Bcl-2. Furthermore, immunofluorescence revealed that SYD can notably decreased the protein level of p53 while reverse the reduction of Claudin-1. CONCLUSIONS These results indicated that radiation enteritis in C57BL/6 mice can be ameliorated by treatment with SYD. The potential protection mechanisms may be involved in ameliorating tissue fibrosis by decreasing inflammatory and apoptotic events.
Collapse
Affiliation(s)
- Zhuohong Li
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Ying Gao
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lei Du
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Ye Yuan
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenbo Huang
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Xi Fu
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yongliang Huang
- Pharmacy Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Xufan Zhang
- Nuclear Medicine Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Shijie Li
- Oncology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
6
|
Quantitative assessment reveals the dominance of duplicated sequences in germline-derived extrachromosomal circular DNA. Proc Natl Acad Sci U S A 2021; 118:2102842118. [PMID: 34789574 PMCID: PMC8617514 DOI: 10.1073/pnas.2102842118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/08/2023] Open
Abstract
Extrachromosomal circular DNA (eccDNA) plays a role in human diseases such as cancer, but little is known about the impact of eccDNA in healthy human biology. Since eccDNA is a tiny fraction of nuclear DNA, artificial amplification has been employed to increase eccDNA amounts, resulting in the loss of native compositions. We developed an approach to enrich eccDNA populations at the native state (naïve small circular DNA, nscDNA) and investigated their origins in the human genome. We found that, in human sperm, the vast majority of nscDNA came from high-copy genomic regions, including the most variable regions between individuals. Because eccDNA can be incorporated back into chromosomes, eccDNA may promote human genetic variation. Extrachromosomal circular DNA (eccDNA) originates from linear chromosomal DNA in various human tissues under physiological and disease conditions. The genomic origins of eccDNA have largely been investigated using in vitro–amplified DNA. However, in vitro amplification obscures quantitative information by skewing the total population stoichiometry. In addition, the analyses have focused on eccDNA stemming from single-copy genomic regions, leaving eccDNA from multicopy regions unexamined. To address these issues, we isolated eccDNA without in vitro amplification (naïve small circular DNA, nscDNA) and assessed the populations quantitatively by integrated genomic, molecular, and cytogenetic approaches. nscDNA of up to tens of kilobases were successfully enriched by our approach and were predominantly derived from multicopy genomic regions including segmental duplications (SDs). SDs, which account for 5% of the human genome and are hotspots for copy number variations, were significantly overrepresented in sperm nscDNA, with three times more sequencing reads derived from SDs than from the entire single-copy regions. SDs were also overrepresented in mouse sperm nscDNA, which we estimated to comprise 0.2% of nuclear DNA. Considering that eccDNA can be integrated into chromosomes, germline-derived nscDNA may be a mediator of genome diversity.
Collapse
|
7
|
Zhang J, Ding N, He Y, Tao C, Liang Z, Xin W, Zhang Q, Wang F. Bioinformatic identification of genomic instability-associated lncRNAs signatures for improving the clinical outcome of cervical cancer by a prognostic model. Sci Rep 2021; 11:20929. [PMID: 34686717 PMCID: PMC8536663 DOI: 10.1038/s41598-021-00384-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
The research is executed to analyze the connection between genomic instability-associated long non-coding RNAs (lncRNAs) and the prognosis of cervical cancer patients. We set a prognostic model up and explored different risk groups' features. The clinical datasets and gene expression profiles of 307 patients have been downloaded from The Cancer Genome Atlas database. We established a prognostic model that combined somatic mutation profiles and lncRNA expression profiles in a tumor genome and identified 35 genomic instability-associated lncRNAs in cervical cancer as a case study. We then stratified patients into low-risk and high-risk groups and were further checked in multiple independent patient cohorts. Patients were separated into two sets: the testing set and the training set. The prognostic model was built using three genomic instability-associated lncRNAs (AC107464.2, MIR100HG, and AP001527.2). Patients in the training set were divided into the high-risk group with shorter overall survival and the low-risk group with longer overall survival (p < 0.001); in the meantime, similar comparable results were found in the testing set (p = 0.046), whole set (p < 0.001). There are also significant differences in patients with histological grades, FIGO stages, and different ages (p < 0.05). The prognostic model focused on genomic instability-associated lncRNAs could predict the prognosis of cervical cancer patients, paving the way for further research into the function and resource of lncRNAs, as well as a key approach to customizing individual care decision-making.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Nan Ding
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Yongxing He
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Chengbin Tao
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhongzhen Liang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wenhu Xin
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qianyun Zhang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Fang Wang
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
8
|
Svetec Miklenić M, Svetec IK. Palindromes in DNA-A Risk for Genome Stability and Implications in Cancer. Int J Mol Sci 2021; 22:2840. [PMID: 33799581 PMCID: PMC7999016 DOI: 10.3390/ijms22062840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
A palindrome in DNA consists of two closely spaced or adjacent inverted repeats. Certain palindromes have important biological functions as parts of various cis-acting elements and protein binding sites. However, many palindromes are known as fragile sites in the genome, sites prone to chromosome breakage which can lead to various genetic rearrangements or even cell death. The ability of certain palindromes to initiate genetic recombination lies in their ability to form secondary structures in DNA which can cause replication stalling and double-strand breaks. Given their recombinogenic nature, it is not surprising that palindromes in the human genome are involved in genetic rearrangements in cancer cells as well as other known recurrent translocations and deletions associated with certain syndromes in humans. Here, we bring an overview of current understanding and knowledge on molecular mechanisms of palindrome recombinogenicity and discuss possible implications of DNA palindromes in carcinogenesis. Furthermore, we overview the data on known palindromic sequences in the human genome and efforts to estimate their number and distribution, as well as underlying mechanisms of genetic rearrangements specific palindromic sequences cause.
Collapse
Affiliation(s)
| | - Ivan Krešimir Svetec
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia;
| |
Collapse
|