Extrachromosomal circular DNA

Last updated

Extrachromosomal circular DNA (eccDNA) is a type of double-stranded circular DNA structure that was first discovered in 1964 by Alix Bassel and Yasuo Hotta. [1] In contrast to previously identified circular DNA structures (e.g., bacterial plasmids, mitochondrial DNA, circular bacterial chromosomes, or chloroplast DNA), eccDNA are circular DNA found in the eukaryotic nuclei of plant and animal (including human) cells. Extrachromosomal circular DNA is derived from chromosomal DNA, can range in size from 50 base pairs to several mega-base pairs in length, and can encode regulatory elements and full-length genes. eccDNA has been observed in various eukaryotic species [2] [3] [4] [5] [6] [7] [8] and it is proposed to be a byproduct of programmed DNA recombination events, such as V(D)J recombination. [8] [9]

Contents

Historical Background

In 1964, Bassel and Hotta published their initial discovery of eccDNA that they made while researching Franklin Stahl’s chromosomal theory. [10] In their experiments, they visualized isolated wheat nuclei and boar sperm by using electron microscopy. [10] Their research found that boar sperm cells contained eccDNA of various sizes. [10] In 1965, Arthur Spriggs’ research group identified eccDNA in the samples of five pediatric patients’ embryonic tumors and one adult patient’s bronchial carcinoma. [11] In the following years, additional research led to the discovery of eccDNA in various species listed in Table 1:

Table 1: Species in which eccDNA has been identified [2]
YearOrganismReference
1965 Boar spermHotta and Bassel, 1965 [10]
1965Human tumorsCox et al., 1965 [11]
1969 Yeast Billheimer and Avers, 1969 [12]
1984 Trypanosomatids Beverly et al., 1984 [13]
1972 Euglena Nass and Ben-Shaul, 1972 [14]
1972 Tobacco Wong and Wildman, 1972 [15]
1972, 1978, 1980FungiAgsteribbe et al., 1972; [16] Stahl et al., 1978; [17] Lazarus et al., 1980 [18]
1972, 1985Cultured human fibroblasts Smith and Vinograd, 1972; [19] Riabowol et al., 1985 [4]
1976 Xenopus Buongiorno-Nardelli et al., 1976 [20]
1978, 1984Chicken bursaDeLap and Rush, 1978; [21] Toda and Yamagishi, 1984 [22]
1982Human tissuesCalabretta et al., 1982 [23]
1983Mouse embryoYamagishi et al., 1983 [24]
1983, 1988, 1990Mouse tissuesTsuda et al., 1983; [25] Flores et al., 1988; [26] Gaubatz and Flores, 1990 [2]
1983Mouse thymocytesYamagishi et al., 1983 [24]
1983Mouse lymphocytesTsuda et al., 1983 [26]

21st Century Research

In the 21st century, researchers have focused on better characterizing the specific subtypes of eccDNA, as well as the structure and function of these molecules within biological systems: [27]

eccDNA Purification

Historically, eccDNA was purified using a two-step procedure that involved first isolating crude extrachromosomal DNA and subsequently digesting linear DNA via exonuclease digestion. [31] Yet, this technique often results in linear DNA contamination because exonuclease digestion is not sufficient to remove all linear DNA. [31] In 2021, Wang et al. developed a three-step eccDNA enrichment method that improved eccDNA purification: [31]

Double minutes (DM) vs. extrachromosomal circular DNA (eccDNA)

Initially, the term double minutes (DM) was commonly used to refer to extrachromosomal circular DNA because it often appeared as a pair in early studies. [27] As research has continued, different subtypes of extrachromosomal circular DNA have been identified that are not double minutes (e.g., microDNA). In 2014, Barreto et al. identified that double minutes only comprise roughly 30% of extrachromosomal DNA. [32] Thus, the term extrachromosomal circular DNA (eccDNA) is becoming more widely used, while the term double minutes is now reserved for a specific subtype of eccDNA. [32]

Structure

eccDNA are circular DNA that have been found in human, plant, and animal cells and are present in the cell nucleus in addition to the chromosomal DNA. eccDNA is distinguishable from other circular DNA in cells, such as mitochondrial DNA (mtDNA), because it ranges in size from a few hundred bases to megabases and is derived from genomic DNA. [1] For example, eccDNA can be formed from exons of protein coding genes, like mucin and titin. Researchers have hypothesized that eccDNA may contribute to the expression of different isoforms of a gene by interfering with or promoting the transcription of specific exons. [1]

eccDNA has been classified as one of four different categories of circular DNA based on size and sequence, including small polydispersed circular DNA (spcDNA), telomeric circles (t-circles), microDNA (100-400 bp), and extrachromosomal DNA (ecDNA). [27] Each of these types has its own unique biological characteristics (see Table 2): [27]

Table 2: Types of eccDNA [27]
Name of eccDNASizeCharacteristicsFunction
spcDNA100–10 kbHighly diverse type of eccDNA, there is a large range of the number of spcDNA found cellsInvolved in human genetic instability
Telomeric circlesmultiples of 738 bpFormed by telomeric arrays, which is a series of repeated sequences at the end of linear DNA.Involved in the alternative lengthening of telomeres (ALT)
microDNA 100-400 bpDerived from genomic locations that have a high GC content and exon densityExpress small functional regulatory RNAs (e.g., microRNAs and new is-like RNAs).
ecDNA 1-3 MbInclude full genes, no telomeres, acentricAmplify genes involved in development of cancer and drug resistance

eccDNA biogenesis

Formation of eccDNA via replication slippage EccDNA formation via replication slippage 2.tif
Formation of eccDNA via replication slippage
The ODERA mechanism of eccDNA formation ODERA Model.tif
The ODERA mechanism of eccDNA formation
EccDNA formation via replication slippage no microdeletion EccDNA formation via replication slippage no microdeletion.tif
EccDNA formation via replication slippage no microdeletion
Double stranded break eccDNA formation Double stranded break eccDNA formation.tif
Double stranded break eccDNA formation

While the exact mechanism for eccDNA generation is still unknown, some studies have suggested that eccDNA generation might be linked to DNA damage repair, [33] hyper-transcription, [33] [34] homologous recombination, [35] and replication stress. [33] There are multiple proposed mechanisms for eccDNA formation: (1) replication slippage creates a loop on the template strand that is then excised and ligated into a circle leaving a microdeletion on the chromosome, (2) replication slippage creates a loop in the product strand that is excised and ligated into a circle that does not generate a microdeletion in the chromosome, (3) the ODERA mechanism of eccDNA formation, and (4) a double stranded break in a repeat region is repaired by homologous recombination, during which the fragment forms a circle and the chromosome suffers a microdeletion [1]

Research conducted in 2021 demonstrated that apoptotic cells are a source of eccDNAs; this was concluded on account of the study showing that apoptotic DNA fragmentation (ADF) is a prerequisite for eccDNA formation through purification methods. [31]

eccDNA can be generated as a result of micro-nuclei formation, indicating chromosomal instability. It has been proposed that premature apoptosis and/or errors in chromosomal segregation during mitosis could lead to micro-nuclei formation. [36]

eccDNA in non-cancerous cells

To test whether eccDNAs occur in non-cancer cells, mouse embryonic stem cells and Southern Blot analysis were used; the results confirmed that eccDNA is found in both cancerous and non-cancerous cells. [31] It is also known that eccDNA is unlikely to be derived from specific genome regions; sequencing data from 2021 reports that the data suggests eccDNAs are widespread across the entirety of the genome. [31] Genome mapping of full-length eccDNAs demonstrated their different genomic alignment patterns, which includes at adjacent, overlapped, or nested positions on the same chromosome or across different chromosomes. [31] eccDNAs originate mostly from single, continuous genomic loci, meaning that one single genomic fragment self-circularizes to form the eccDNA, rather than being formed from ligation of different genomic fragments. [31] These two variants can be classified as continuous and non-continuous eccDNAs, respectively. [31] To further understand the reason behind the circularization of fragmented DNA, the three various mammalian ligase enzymes were tested: Lig1, Lig3, and Lig4 [31] . Using knockout models in the CH12F3 mouse B-lymphocyte cell line, research conducted in 2021 identified Lig3 as the main ligase for eccDNA generation in these cells. [31]

Function

The exact function of eccDNA has been debated, but some studies have suggested that eccDNAs might contribute to gene amplification in cancer, [1] immune function, [31] and aging. [34] [35] [37]

eccDNA function in immune system

According to research conducted in 2021, another function of eccDNAs is their role as possible immunostimulants. [31] eccDNA significantly induces type I interferons (IFNα, IFNβ), interleukin-6 (IL-6), and tumor necrosis factor (TNF), even more so than linear DNA and other generally potent cytokine inducers at their highest concentration levels. [31] Similar patterns are observed with macrophages as the data showed that eccDNAs are very potent immunostimulants in activating both bone marrow-derived dendritic cells and bone marrow-derived macrophages. [31] Additionally, experiments altered the eccDNA structure with one nick per eccDNA segment and subsequently treated with enzymes to generate linear versions of the eccDNA. [31] In these experiments, cytokine transcription, an important marker for immune system activity, was shown to be much higher in the non-treated eccDNA compared to the linearized treatment, conferring that the circular structure of eccDNA rather than the genetic sequence itself gives the eccDNA its immune function. [31]

eccDNA function in cancer

Some known functions of eccDNA include contributions to intercellular genetic heterogeneity in tumors, and more specifically the amplification of oncogenes and drug-resistant genes. This also supports that the genes on eccDNA are expressed. Overall, eccDNA has been linked to cancer and drug resistance, aging, gene compensation, [1] and for this reason it continues to be a significant topic of discussion.

Applications

Role in cancer

A subtype of eccDNA, such as ecDNA, ribosomal DNA locus (Extrachromosomal rDNA circle), and double minutes have been associated with genomic instability. Double minute ecDNAs are fragments of extrachromosomal DNA, which were originally observed in a large number of human tumors including breast, lung, ovary, colon, and most notably, neuroblastoma. They are a manifestation of gene amplification during the development of tumors, which give the cells selective advantages for growth and survival. Double minutes, like actual chromosomes, are composed of chromatin and replicate in the nucleus of the cell during cell division. Unlike typical chromosomes, they are composed of circular fragments of DNA, up to only a few million base pairs in size and contain no centromere or telomere.

Double minute chromosomes (DMs), which present as paired chromatin bodies under light microscopy, have been shown to be a subset of ecDNA. [28] [38] Double minute chromosomes represent about 30% of the cancer-containing spectrum of ecDNA, including single bodies, [28] and have been found to contain identical gene content as single bodies. The ecDNA notation encompasses all forms of the large gene-containing extrachromosomal DNA found in cancer cells. This type of ecDNA is commonly seen in cancer cells of various histologies, but virtually never in normal tissue. [39] [28] ecDNA are thought to be produced through double-strand breaks in chromosomes or over replication of DNA in an organism. [40]

The circular shape of ecDNA differs from the linear structure of chromosomal DNA in meaningful ways that influence cancer pathogenesis. [41] [30] Oncogenes encoded on ecDNA have massive transcriptional output, ranking in the top 1% of genes in the entire transcriptome. In contrast to bacterial plasmids or mitochondrial DNA, ecDNA are chromatinized, containing high levels of active histone marks, but a paucity of repressive histone marks. The ecDNA chromatin architecture lacks the higher-order compaction that is present on chromosomal DNA and is among the most accessible DNA in the entire cancer genome.

From eccDNA, matrix attachment regions (MARs) were found to activate amplification of oncogenes. [1] Transfection of these MARs into human embryonic kidney 293T cells resulted in an increase in gene expression, suggesting that these eccDNA-derived MARs are involved in oncogene activation. [42] eccDNA also appears to play a role in other cancers such as breast cancer, where oncogenes in human epidermal growth factor receptor 2 (HER2)-positive breast cancer genes in eccDNA are amplified. [1] This eccDNA has also shown the ability to acquire resistance to therapies for receptor tyrosine kinases (RTKs), like HER26. [43]

Role in aging

Yeast are model organisms for studying aging, and eccDNAs have been shown to accumulate in old cells and play a role in causing aging in yeast. [37] Speculation continues on the generality of this concept in higher species, like mammals. [37]

See also

Related Research Articles

<span class="mw-page-title-main">Plasmid</span> Small DNA molecule within a cell

A plasmid is a small, extrachromosomal DNA molecule within a cell that is physically separated from chromosomal DNA and can replicate independently. They are most commonly found as small circular, double-stranded DNA molecules in bacteria; however, plasmids are sometimes present in archaea and eukaryotic organisms. In nature, plasmids often carry genes that benefit the survival of the organism and confer selective advantage such as antibiotic resistance. While chromosomes are large and contain all the essential genetic information for living under normal conditions, plasmids are usually very small and contain only additional genes that may be useful in certain situations or conditions. Artificial plasmids are widely used as vectors in molecular cloning, serving to drive the replication of recombinant DNA sequences within host organisms. In the laboratory, plasmids may be introduced into a cell via transformation. Synthetic plasmids are available for procurement over the internet.

A microsatellite is a tract of repetitive DNA in which certain DNA motifs are repeated, typically 5–50 times. Microsatellites occur at thousands of locations within an organism's genome. They have a higher mutation rate than other areas of DNA leading to high genetic diversity. Microsatellites are often referred to as short tandem repeats (STRs) by forensic geneticists and in genetic genealogy, or as simple sequence repeats (SSRs) by plant geneticists.

<span class="mw-page-title-main">Yeast artificial chromosome</span> Genetically engineered chromosome derived from the DNA of yeast

Yeast artificial chromosomes (YACs) are genetically engineered chromosomes derived from the DNA of the yeast, Saccharomyces cerevisiae, which is then ligated into a bacterial plasmid. By inserting large fragments of DNA, from 100–1000 kb, the inserted sequences can be cloned and physically mapped using a process called chromosome walking. This is the process that was initially used for the Human Genome Project, however due to stability issues, YACs were abandoned for the use of bacterial artificial chromosome

In molecular biology, an amplicon is a piece of DNA or RNA that is the source and/or product of amplification or replication events. It can be formed artificially, using various methods including polymerase chain reactions (PCR) or ligase chain reactions (LCR), or naturally through gene duplication. In this context, amplification refers to the production of one or more copies of a genetic fragment or target sequence, specifically the amplicon. As it refers to the product of an amplification reaction, amplicon is used interchangeably with common laboratory terms, such as "PCR product."

Double minutes (DMs) are small fragments of extrachromosomal DNA, which have been observed in a large number of human tumors including breast, lung, ovary, colon, and most notably, neuroblastoma. They are a manifestation of gene amplification as a result of chromothripsis, during the development of tumors, which give the cells selective advantages for growth and survival. This selective advantage is as a result of double minutes frequently harboring amplified oncogenes and genes involved in drug resistance. DMs, like actual chromosomes, are composed of chromatin and replicate in the nucleus of the cell during cell division. Unlike typical chromosomes, they are composed of circular fragments of DNA, up to only a few million base pairs in size, and contain no centromere or telomere. Further to this, they often lack key regulatory elements, allowing genes to be constitutively expressed. The term ecDNA may be used to refer to DMs in a more general manner. The term Double Minute originates from the visualization of these features under microscope; double because the dots were found in pairs, and minute because they were minuscule.

Carcinogenesis, also called oncogenesis or tumorigenesis, is the formation of a cancer, whereby normal cells are transformed into cancer cells. The process is characterized by changes at the cellular, genetic, and epigenetic levels and abnormal cell division. Cell division is a physiological process that occurs in almost all tissues and under a variety of circumstances. Normally, the balance between proliferation and programmed cell death, in the form of apoptosis, is maintained to ensure the integrity of tissues and organs. According to the prevailing accepted theory of carcinogenesis, the somatic mutation theory, mutations in DNA and epimutations that lead to cancer disrupt these orderly processes by interfering with the programming regulating the processes, upsetting the normal balance between proliferation and cell death. This results in uncontrolled cell division and the evolution of those cells by natural selection in the body. Only certain mutations lead to cancer whereas the majority of mutations do not.

<span class="mw-page-title-main">Rolling circle replication</span> DNA synthesis technique

Rolling circle replication (RCR) is a process of unidirectional nucleic acid replication that can rapidly synthesize multiple copies of circular molecules of DNA or RNA, such as plasmids, the genomes of bacteriophages, and the circular RNA genome of viroids. Some eukaryotic viruses also replicate their DNA or RNA via the rolling circle mechanism.

Extrachromosomal DNA is any DNA that is found off the chromosomes, either inside or outside the nucleus of a cell. Most DNA in an individual genome is found in chromosomes contained in the nucleus. Multiple forms of extrachromosomal DNA exist, and, while some of these serve important biological functions, they can also play a role in diseases such as cancer.

Myc is a family of regulator genes and proto-oncogenes that code for transcription factors. The Myc family consists of three related human genes: c-myc (MYC), l-myc (MYCL), and n-myc (MYCN). c-myc was the first gene to be discovered in this family, due to homology with the viral gene v-myc.

<span class="mw-page-title-main">HMGA2</span> Protein-coding gene in the species Homo sapiens

High-mobility group AT-hook 2, also known as HMGA2, is a protein that, in humans, is encoded by the HMGA2 gene.

<span class="mw-page-title-main">UBE2C</span> Protein-coding gene in the species Homo sapiens

Ubiquitin-conjugating enzyme E2 C is a protein that in humans is encoded by the UBE2C gene.

<span class="mw-page-title-main">TSPAN31</span> Protein-coding gene in humans

Tetraspanin-31 is a protein that in humans is encoded by the TSPAN31 gene.

<span class="mw-page-title-main">EIF5A2</span> Protein-coding gene in the species Homo sapiens

Eukaryotic translation initiation factor 5A-2 is a protein that in humans is encoded by the EIF5A2 gene.

<span class="mw-page-title-main">BCAS1</span> Protein-coding gene in the species Homo sapiens

Breast carcinoma-amplified sequence 1 is a protein that in humans is encoded by the BCAS1 gene.

<span class="mw-page-title-main">MIRH1</span>

Putative microRNA host gene 1 protein is a protein that in humans is encoded by the MIR17HG gene.

Extrachromosomal rDNA circles are pieces of extrachromosomal circular DNA (eccDNA) derived from ribosomal DNA (rDNA). Initially found in baker's yeast, these self-replicating circles are suggested to contribute to their aging and found in their aged cells. Like ordinary eccDNA, they are created by intra-molecular homologous recombination of the chromosome. The process for intra-molecular homologous recombination is independent of chromosomal replication. The de novo generated circles had exact multiples of tandem copies of 2-kb fragments from cosmid templates. The tandem organization is essential to circle formation. Looping out of organized ribosomal genes in intergenic nontranscribed spacers yielded either large or small repeat circles dependent on large or short repeats of the spacer.

<span class="mw-page-title-main">Chromothripsis</span> Massive chromosomal rearrangement process linked to cancer

Chromothripsis is a mutational process by which up to thousands of clustered chromosomal rearrangements occur in a single event in localised and confined genomic regions in one or a few chromosomes, and is known to be involved in both cancer and congenital diseases. It occurs through one massive genomic rearrangement during a single catastrophic event in the cell's history. It is believed that for the cell to be able to withstand such a destructive event, the occurrence of such an event must be the upper limit of what a cell can tolerate and survive. The chromothripsis phenomenon opposes the conventional theory that cancer is the gradual acquisition of genomic rearrangements and somatic mutations over time.

<span class="mw-page-title-main">MicroDNA</span>

MicroDNA is the most abundant subtype of Extrachromosomal Circular DNA (eccDNA) in humans, typically ranging from 200-400 base pairs in length and enriched in non-repetitive genomic sequences with a high density of exons. Additionally, microDNA has been found to come from regions with CpG-islands which are commonly found within the 5' and 3' UTRs. Being produced from regions of active transcription, it is hypothesized that microDNA may be formed as a by-product of transcriptional DNA damage repair. MicroDNA is also thought to arise from other DNA repair pathways, mainly due to the parental sequences of microDNA having 2- to 15 bp direct repeats at the ends, resulting in replication slippage repair. While only recently discovered, the role microDNA plays in and out of the cell is still not completely understood. However, microDNA is currently thought to affect cellular homeostasis through transcription factor binding and have been used as a cancer biomarker.

Paul S. Mischel is an American physician-scientist whose laboratory has made pioneering discoveries in the pathogenesis of human cancer. He is currently a Professor and Vice Chair of Research for the Department of Pathology and Institute Scholar of ChEM-H, Stanford University. Mischel was elected into the American Society for Clinical Investigation (ASCI), serving as ASCI president in 2010/11. He was inducted into the Association of American Physicians, and was elected as a fellow of the American Association for the Advancement of Science.

Anindya Dutta is an Indian-born American biochemist and cancer researcher, a Chair of the Department of Genetics at the University of Alabama at Birmingham School of Medicine since 2021, who has served as Chair of the Department of Biochemistry and Molecular Genetics at the University of Virginia School of Medicine in 2011–2021. Dutta's research has focused on the mammalian cell cycle with an emphasis on DNA replication and repair and on noncoding RNAs. He is particularly interested in how de-regulation of these processes promote cancer progression. For his accomplishments he has been elected a Fellow of the American Association for the Advancement of Science, received the Ranbaxy Award in Biomedical Sciences, the Outstanding Investigator Award from the American Society for Investigative Pathology, the Distinguished Scientist Award from the University of Virginia and the Mark Brothers Award from the Indiana University School of Medicine.

References

  1. 1 2 3 4 5 6 7 8 Paulsen T, Kumar P, Koseoglu MM, Dutta A (April 2018). "Discoveries of Extrachromosomal Circles of DNA in Normal and Tumor Cells". Trends in Genetics. 34 (4): 270–278. doi:10.1016/j.tig.2017.12.010. PMC   5881399 . PMID   29329720.
  2. 1 2 3 Gaubatz JW (1990). "Extrachromosomal circular DNAs and genomic sequence plasticity in eukaryotic cells". Mutation Research. 237 (5–6): 271–292. doi:10.1016/0921-8734(90)90009-g. PMID   2079966.
  3. Cohen S, Yacobi K, Segal D (June 2003). "Extrachromosomal circular DNA of tandemly repeated genomic sequences in Drosophila". Genome Research. 13 (6A): 1133–1145. doi:10.1101/gr.907603. PMC   403641 . PMID   12799349.
  4. 1 2 Cohen S, Agmon N, Sobol O, Segal D (March 2010). "Extrachromosomal circles of satellite repeats and 5S ribosomal DNA in human cells". Mobile DNA. 1 (1): 11. doi: 10.1186/1759-8753-1-11 . PMC   3225859 . PMID   20226008.
  5. Stanfield S, Helinski DR (October 1976). "Small circular DNA in Drosophila melanogaster". Cell. 9 (2): 333–345. doi:10.1016/0092-8674(76)90123-9. PMID   824055. S2CID   39382051.
  6. 1 2 3 Shibata Y, Kumar P, Layer R, Willcox S, Gagan JR, Griffith JD, Dutta A (April 2012). "Extrachromosomal microDNAs and chromosomal microdeletions in normal tissues". Science. 336 (6077): 82–86. Bibcode:2012Sci...336...82S. doi:10.1126/science.1213307. PMC   3703515 . PMID   22403181.
  7. Møller HD, Parsons L, Jørgensen TS, Botstein D, Regenberg B (June 2015). "Extrachromosomal circular DNA is common in yeast". Proceedings of the National Academy of Sciences of the United States of America. 112 (24): E3114–E3122. Bibcode:2015PNAS..112E3114M. doi: 10.1073/pnas.1508825112 . PMC   4475933 . PMID   26038577.
  8. 1 2 Shoura MJ, Gabdank I, Hansen L, Merker J, Gotlib J, Levene SD, Fire AZ (October 2017). "Intricate and Cell Type-Specific Populations of Endogenous Circular DNA (eccDNA) in Caenorhabditis elegans and Homo sapiens". G3. 7 (10): 3295–3303. doi:10.1534/g3.117.300141. PMC   5633380 . PMID   28801508.
  9. Hayday AC, Saito H, Gillies SD, Kranz DM, Tanigawa G, Eisen HN, Tonegawa S (February 1985). "Structure, organization, and somatic rearrangement of T cell gamma genes". Cell. 40 (2): 259–269. doi:10.1016/0092-8674(85)90140-0. PMID   3917858. S2CID   34582929.
  10. 1 2 3 4 Hotta Y, Bassel A (February 1965). "Molecular Size and Circularity of DNA in Cells of Mammals and Higher Plants". Proceedings of the National Academy of Sciences of the United States of America. 53 (2): 356–362. Bibcode:1965PNAS...53..356H. doi: 10.1073/pnas.53.2.356 . PMC   219520 . PMID   14294069.
  11. 1 2 Cox D, Yuncken C, Spriggs AI (July 1965). "Minute Chromatin Bodies in Malignant Tumours of Childhood". Lancet. 1 (7402): 55–58. doi:10.1016/s0140-6736(65)90131-5. PMID   14304929.
  12. Billheimer FE, Avers CJ (October 1969). "Nuclear and mitochondrial DNA from wild-type and petite yeast: circularity, length, and buoyant density". Proceedings of the National Academy of Sciences of the United States of America. 64 (2): 739–746. Bibcode:1969PNAS...64..739B. doi: 10.1073/pnas.64.2.739 . PMC   223406 . PMID   5261045.
  13. Beverley SM, Coderre JA, Santi DV, Schimke RT (September 1984). "Unstable DNA amplifications in methotrexate-resistant Leishmania consist of extrachromosomal circles which relocalize during stabilization". Cell. 38 (2): 431–439. doi:10.1016/0092-8674(84)90498-7. PMID   6467372. S2CID   2030494.
  14. Nass MM, Ben-Shaul Y (June 1972). "A novel closed circular duplex DNA in bleached mutant and green strains of Euglena gracilis". Biochimica et Biophysica Acta (BBA) - Nucleic Acids and Protein Synthesis. 272 (1): 130–136. doi:10.1016/0005-2787(72)90041-X. PMID   4625469.
  15. Wong FY, Wildman SG (January 1972). "Simple procedure for isolation of satellite DNA's from tobacco leaves in high yield and demonstration of minicircles". Biochimica et Biophysica Acta (BBA) - Nucleic Acids and Protein Synthesis. 259 (1): 5–12. doi:10.1016/0005-2787(72)90468-6. PMID   5011974.
  16. Agsteribbe E, Kroon AM, van Bruggen EF (May 1972). "Circular DNA from mitochondria of Neurospora crassa". Biochimica et Biophysica Acta (BBA) - Nucleic Acids and Protein Synthesis. 269 (2): 299–303. doi:10.1016/0005-2787(72)90439-X. PMID   4260513.
  17. Stahl U, Lemke PA, Tudzynski P, Kück U, Esser K (July 1978). "Evidence for plasmid like DNA in a filamentous fungus, the ascomycete Podospora anserina". Molecular & General Genetics. 162 (3): 341–343. doi:10.1007/BF00268860. PMID   683172. S2CID   12655440.
  18. Lazarus CM, Earl AJ, Turner G, Küntzel H (May 1980). "Amplification of a mitochondrial DNA sequence in the cytoplasmically inherited 'ragged' mutant of Aspergillus amstelodami". European Journal of Biochemistry. 106 (2): 633–641. doi: 10.1111/j.1432-1033.1980.tb04611.x . PMID   6249580.
  19. Smith CA, Vinograd J (August 1972). "Small polydisperse circular DNA of HeLa cells". Journal of Molecular Biology. 69 (2): 163–178. doi:10.1016/0022-2836(72)90222-7. PMID   5070865.
  20. Buongiorno-Nardelli M, Amaldi F, Lava-Sanchez PA (March 1976). "Electron microscope analysis of amplifying ribosomal DNA from Xenopus laevis". Experimental Cell Research. 98 (1): 95–103. doi:10.1016/0014-4827(76)90467-5. PMID   1253845.
  21. DeLap RJ, Rush MG (December 1978). "Change in quantity and size distribution of small circular DNAs during development of chicken bursa". Proceedings of the National Academy of Sciences of the United States of America. 75 (12): 5855–5859. Bibcode:1978PNAS...75.5855D. doi: 10.1073/pnas.75.12.5855 . PMC   393074 . PMID   282606.
  22. Toda M, Hirama T, Takeshita S, Yamagishi H (June 1989). "Excision products of immunoglobulin gene rearrangements". Immunology Letters. 21 (4): 311–316. doi:10.1016/0165-2478(89)90025-4. PMID   2767726.
  23. Calabretta B, Robberson DL, Barrera-Saldaña HA, Lambrou TP, Saunders GF (March 1982). "Genome instability in a region of human DNA enriched in Alu repeat sequences". Nature. 296 (5854): 219–225. Bibcode:1982Natur.296..219C. doi:10.1038/296219a0. PMID   6278320. S2CID   4265874.
  24. 1 2 Yamagishi H, Kunisada T, Iwakura Y, Nishimune Y, Ogiso Y, Matsushiro A (December 1983). "Emergence of the Extrachromosomal Circular DNA Complexes as One of the Earliest Signals of Cellular Differentiation in the Early Development of Mouse Embryo. (mouse embryo/teratocarcinoma/mica-press-adsorption/circular DNA complex/DNA rearrangement)". Development, Growth and Differentiation. 25 (6): 563–569. doi: 10.1111/j.1440-169X.1983.00563.x . PMID   37282129. S2CID   83666742.
  25. Tsuda T, Yamagishi H, Ohnishi N, Yamada Y, Izumi H, Mori KJ (November 1983). "Extrachromosomal circular DNAs from murine hemopoietic tissue cells". Plasmid. 10 (3): 235–241. doi:10.1016/0147-619X(83)90037-9. PMID   6657775.
  26. 1 2 Flores SC, Sunnerhagen P, Moore TK, Gaubatz JW (May 1988). "Characterization of repetitive sequence families in mouse heart small polydisperse circular DNAs: age-related studies". Nucleic Acids Research. 16 (9): 3889–3906. doi:10.1093/nar/16.9.3889. PMC   336563 . PMID   3375074.
  27. 1 2 3 4 5 Wang T, Zhang H, Zhou Y, Shi J (June 2021). "Extrachromosomal circular DNA: a new potential role in cancer progression". Journal of Translational Medicine. 19 (1): 257. doi: 10.1186/s12967-021-02927-x . PMC   8194206 . PMID   34112178.
  28. 1 2 3 4 5 6 Turner KM, Deshpande V, Beyter D, Koga T, Rusert J, Lee C, et al. (March 2017). "Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity". Nature. 543 (7643): 122–125. Bibcode:2017Natur.543..122T. doi:10.1038/nature21356. PMC   5334176 . PMID   28178237.
  29. Møller HD, Mohiyuddin M, Prada-Luengo I, Sailani MR, Halling JF, Plomgaard P, et al. (March 2018). "Circular DNA elements of chromosomal origin are common in healthy human somatic tissue". Nature Communications. 9 (1): 1069. Bibcode:2018NatCo...9.1069M. doi:10.1038/s41467-018-03369-8. PMC   5852086 . PMID   29540679.
  30. 1 2 Wu S, Turner KM, Nguyen N, Raviram R, Erb M, Santini J, et al. (November 2019). "Circular ecDNA promotes accessible chromatin and high oncogene expression". Nature. 575 (7784): 699–703. Bibcode:2019Natur.575..699W. doi:10.1038/s41586-019-1763-5. PMC   7094777 . PMID   31748743.
  31. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Wang Y, Wang M, Djekidel MN, Chen H, Liu D, Alt FW, Zhang Y (November 2021). "eccDNAs are apoptotic products with high innate immunostimulatory activity". Nature. 599 (7884): 308–314. Bibcode:2021Natur.599..308W. doi: 10.1038/s41586-021-04009-w . PMC   9295135 . PMID   34671165. S2CID   239051756.
  32. 1 2 Barreto SC, Uppalapati M, Ray A (May 2014). "Small Circular DNAs in Human Pathology". The Malaysian Journal of Medical Sciences. 21 (3): 4–18. PMC   4163554 . PMID   25246831.
  33. 1 2 3 Dillon LW, Kumar P, Shibata Y, Wang YH, Willcox S, Griffith JD, et al. (June 2015). "Production of Extrachromosomal MicroDNAs Is Linked to Mismatch Repair Pathways and Transcriptional Activity". Cell Reports. 11 (11): 1749–1759. doi:10.1016/j.celrep.2015.05.020. PMC   4481157 . PMID   26051933.
  34. 1 2 Hull RM, King M, Pizza G, Krueger F, Vergara X, Houseley J (December 2019). "Transcription-induced formation of extrachromosomal DNA during yeast ageing". PLOS Biology. 17 (12): e3000471. doi: 10.1371/journal.pbio.3000471 . PMC   6890164 . PMID   31794573.
  35. 1 2 Gresham D, Usaite R, Germann SM, Lisby M, Botstein D, Regenberg B (October 2010). "Adaptation to diverse nitrogen-limited environments by deletion or extrachromosomal element formation of the GAP1 locus". Proceedings of the National Academy of Sciences of the United States of America. 107 (43): 18551–18556. Bibcode:2010PNAS..10718551G. doi: 10.1073/pnas.1014023107 . PMC   2972935 . PMID   20937885.
  36. Altungöz; Yüksel (September 2023). "Gene amplifications and extrachromosomal circular DNAs: function and biogenesis". Molecular Biology Reports. 50 (9): 7693–7703. doi:10.1007/s11033-023-08649-1. PMID   37433908 via Springer.
  37. 1 2 3 Sinclair DA, Guarente L (December 1997). "Extrachromosomal rDNA circles--a cause of aging in yeast". Cell. 91 (7): 1033–1042. doi: 10.1016/s0092-8674(00)80493-6 . PMID   9428525. S2CID   12735979.
  38. deCarvalho AC, Kim H, Poisson LM, Winn ME, Mueller C, Cherba D, et al. (May 2018). "Discordant inheritance of chromosomal and extrachromosomal DNA elements contributes to dynamic disease evolution in glioblastoma". Nature Genetics. 50 (5): 708–717. doi:10.1038/s41588-018-0105-0. PMC   5934307 . PMID   29686388.
  39. Kim H, Nguyen NP, Turner K, Wu S, Gujar AD, Luebeck J, et al. (September 2020). "Extrachromosomal DNA is associated with oncogene amplification and poor outcome across multiple cancers". Nature Genetics. 52 (9): 891–897. doi:10.1038/s41588-020-0678-2. PMC   7484012 . PMID   32807987.
  40. Kuttler F, Mai S (February 2007). "Formation of non-random extrachromosomal elements during development, differentiation and oncogenesis". Seminars in Cancer Biology. 17 (1): 56–64. doi:10.1016/j.semcancer.2006.10.007. PMID   17116402.
  41. Zimmer C (November 20, 2019). "Scientists Are Just Beginning to Understand Mysterious DNA Circles Common in Cancer Cells". New York Times.
  42. Jin Y, Liu Z, Cao W, Ma X, Fan Y, Yu Y, et al. (2012). "Novel functional MAR elements of double minute chromosomes in human ovarian cells capable of enhancing gene expression". PLOS ONE. 7 (2): e30419. Bibcode:2012PLoSO...730419J. doi: 10.1371/journal.pone.0030419 . PMC   3272018 . PMID   22319568.
  43. Vicario R, Peg V, Morancho B, Zacarias-Fluck M, Zhang J, Martínez-Barriocanal Á, et al. (2015-06-15). "Patterns of HER2 Gene Amplification and Response to Anti-HER2 Therapies". PLOS ONE. 10 (6): e0129876. Bibcode:2015PLoSO..1029876V. doi: 10.1371/journal.pone.0129876 . PMC   4467984 . PMID   26075403.

Further reading