Fact-checked by Grok 2 weeks ago

Extrachromosomal DNA

Extrachromosomal DNA (ecDNA), also referred to as extrachromosomal circular DNA (eccDNA), consists of circular, double-stranded DNA molecules that exist independently of the chromosomal genome in cells, including both prokaryotes (such as plasmids) and eukaryotes, typically ranging in size from dozens of base pairs to several megabases. First identified in bacteria as plasmids in the 1950s and later in eukaryotic cancer cells as double-minute chromosomes in the 1960s, these structures are found in both normal and diseased tissues but are particularly abundant in cancer cells, where they often contain amplified oncogenes and regulatory elements, driving rapid tumor evolution and heterogeneity. Unlike linear chromosomal DNA, ecDNA lacks centromeres and telomeres, enabling non-Mendelian inheritance patterns that facilitate high copy number variation and genetic instability. The biogenesis of ecDNA involves multiple mechanisms, including chromosomal breakage events such as , breakage-fusion-bridge cycles, and replication fork stalling, which generate acentric circular fragments that are subsequently amplified. Structurally, ecDNA exhibits highly accessible , allowing for enhanced transcriptional activity of contained , and can form dynamic hubs that interact with chromosomal DNA to co-regulate expression. In normal cells, small eccDNAs (e.g., microDNAs) may arise from gene excision and contribute to plasticity or maintenance, but in cancer, larger forms predominate and are often derived from a single arm. In , ecDNA plays a pivotal role in tumorigenesis by amplifying key drivers like , , and , promoting aggressive phenotypes, , and immune evasion through altered . Detected in approximately 17% of tumor samples across various cancers, including high prevalence in glioblastomas (up to 49%) and liposarcomas (up to 55%), ecDNA is strongly associated with poor , with patients showing reduced survival (hazard ratio 1.44) and increased treatment resistance to therapies like and targeted inhibitors. Emerging as a , ecDNA can be identified in from plasma, offering potential for non-invasive diagnostics, while therapeutic strategies targeting its loss—such as hydroxyurea or —hold promise for overcoming resistance.

Overview

Definition and Characteristics

Extrachromosomal DNA (ecDNA), often specifically referring to extrachromosomal circular DNA (eccDNA), consists of DNA molecules that exist independently of the primary chromosomal complement within a , typically in circular form and not integrated into the main . In contrast to chromosomal DNA, which is organized into linear chromosomes within the of eukaryotes or the of prokaryotes, ecDNA can reside either within the as separate entities or extranuclearly (e.g., in organelles), often adopting a circular to support autonomous replication without reliance on chromosomal machinery. This independence allows ecDNA to propagate separately, though it typically lacks centromeric sequences in nuclear contexts, resulting in unstable inheritance during due to random rather than equitable distribution. Key characteristics of ecDNA include its relatively small size, ranging from hundreds of base pairs to several megabases ( to ), which facilitates high copy numbers and rapid amplification compared to the larger chromosomal genomes. It possesses the potential for autonomous replication through incorporated origins of replication, enabling persistence across generations without integration, and is prevalent in diverse biological contexts across prokaryotes and eukaryotes. While predominantly circular—enhancing stability and replication efficiency—linear extrachromosomal elements, such as double-minute chromosomes in cancer cells, also occur and contribute to . Representative examples of ecDNA include plasmids in prokaryotes, which serve as vehicles for resistance and metabolic functions; (mtDNA) in eukaryotic organelles, encoding essential respiratory proteins; viral episomes, such as those from herpesviruses that maintain ; and cancer-associated circular ecDNA, often amplifying oncogenes like or to drive tumor progression. EcDNA is ubiquitous in , from bacterial populations to pathologies, with detection in up to 17% of tumor samples across various cancer types, underscoring its role in normal and .

Historical Discovery

The concept of extrachromosomal DNA emerged from early 20th-century experiments demonstrating genetic transformation in bacteria. In 1928, Frederick Griffith observed that heat-killed smooth pneumococci could transfer virulence traits to non-virulent rough strains in mice, providing the first evidence of a heritable factor independent of the bacterial chromosome. This laid the groundwork for understanding non-chromosomal genetic elements, later identified as plasmids. The 1940s and 1950s brought key breakthroughs in bacterial genetics, solidifying the existence of extrachromosomal DNA. Joshua Lederberg and Edward Tatum's 1946 experiments revealed genetic recombination in Escherichia coli through conjugation, indicating the transfer of extrachromosomal factors between cells. In 1952, Norton Zinder and Lederberg discovered transduction, where bacteriophages mediate the transfer of bacterial DNA fragments, further highlighting mobile genetic elements. That same year, Lederberg coined the term "plasmid" to describe these self-replicating, extrachromosomal DNA molecules, linking them to traits like antibiotic resistance observed in clinical isolates. In eukaryotes, the discovery of extrachromosomal DNA in organelles marked significant milestones in the 1960s. Margit M. K. Nass and Sylvan Nass identified DNA within mitochondria using electron microscopy in 1963, visualizing DNase-sensitive fibers distinct from nuclear DNA. For chloroplasts, biochemical evidence of DNA appeared in 1959 through incorporation of into Spirogyra chloroplasts by Ralph Stocking and Ernest Gifford, with electron microscopic confirmation in 1962 by Hans Ris and Walther Plaut, establishing plastid autonomy. By the , these findings supported the endosymbiotic theory, recognizing organellar genomes as separate from nuclear chromosomes. In the cancer context, extrachromosomal DNA was first observed as double-minute chromosomes in tumor cells during the . A. I. Spriggs and colleagues reported these small, paired bodies in a human pleural effusion sample in 1962, initially without recognizing their DNA . By the 1980s, double minutes were linked to in tumors, serving as precursors to circular extrachromosomal DNA (ecDNA). The 2010s revived interest through next-generation sequencing (NGS), with a 2020 study confirming circular ecDNA in , driving amplification and genome remodeling. Post-2020 research, including a 2024 , has underscored ecDNA's role in treatment resistance across cancers, amplifying its genomic impact.

Biogenesis

Mechanisms of Formation

Extrachromosomal DNA (ecDNA) arises through several molecular processes that disrupt chromosomal , primarily involving the excision or misrepair of genomic segments. Key mechanisms include chromosomal excision, replication errors, and failures in double-strand break repair, each contributing to the release of non-integrated DNA elements that can persist extrachromosomally. These pathways often intersect with broader genomic instability, enabling the formation of circular ecDNA structures. Chromosomal excision typically occurs via microhomology-mediated deletion (MMEJ), where short homologous sequences (1–20 bp) at DNA break sites facilitate the deletion of intervening segments, releasing acentric DNA fragments that can circularize. This process is error-prone and generates small ecDNA molecules, often termed microDNA, without leaving prominent chromosomal scars. Replication errors, such as fork stalling and template switching (FoSTeS), further promote ecDNA formation during S-phase; stalled replication forks switch to alternative templates, leading to the excision of looped-out DNA segments that form extrachromosomal circles via or . These mechanisms are particularly active in contexts of , allowing rapid generation of ecDNA from amplified regions. Failures in double-strand break repair play a central role in ecDNA biogenesis, with (NHEJ) or alternative end joining (alt-EJ) pathways ligating broken DNA ends to form circular structures. In NHEJ, Ku proteins and DNA-PK recruit ligases to join incompatible ends, often resulting in small deletions, while alt-EJ relies on microhomologies for more precise but still mutagenic circularization. Genomic instability amplifies these processes; for instance, within micronuclei—small, extranuclear compartments formed from chromosome fragments—shatters DNA into pieces that reassemble into complex ecDNA via erroneous repair. Circular forms predominate due to head-to-tail ligation of multiple fragments, stabilizing the molecule against degradation. Recent studies have identified additional mechanisms, such as YY1-mediated DNA looping that facilitates religation by the Lig3-YY1 complex, further contributing to ecDNA biogenesis. Several factors enhance ecDNA formation, including defects in DNA damage response pathways such as /2 mutations, which impair and favor error-prone NHEJ, increasing ecDNA prevalence. Similarly, replication stress induced by activation (e.g., overexpression) causes fork collapse and DSBs, promoting excision events. Quantitatively, ecDNA amplifies rapidly post-formation, achieving high copy numbers—often tens to hundreds per cell—through activation of multiple extrareplicative origins, far exceeding chromosomal duplication rates and enabling swift increases. Depletion of classical NHEJ-promoting components, like 53BP1, increases ecDNA levels by favoring alt-EJ and MMEJ pathways, underscoring the role of error-prone repair in ecDNA biogenesis.

Structural Properties

Extrachromosomal DNA (ecDNA) predominantly adopts a circular, double-stranded , which provides and allows for independent replication outside of conventional chromosomes. These circular forms typically incorporate origins of replication and promoter sequences, enabling autonomous propagation within the . The size of ecDNA molecules spans a broad spectrum, ranging from approximately 100 base pairs for small circular DNAs to more than 1 megabase for larger entities often termed ecDNA in cancer contexts. These structures frequently harbor functional gene clusters, including oncogenes like , , or FGFR2, along with associated regulatory elements such as enhancers. Circular ecDNA generally does not require telomeric caps, while centromeric sequences appear variably, though functional centromeres are rare, contributing to patterns. ecDNA associates with proteins to form -based structures, albeit with distinct organizational features compared to linear chromosomes. These associations include binding by canonical s, facilitating a chromatin-like architecture that supports regulatory functions. Epigenetically, ecDNA displays heightened accessibility and pronounced enrichment in active marks, particularly H3K27ac, which correlates with enhanced promoter and enhancer activity. This open configuration often results in the absence of repressive marks and incomplete occupancy, promoting rapid transcriptional activation and elevated relative to chromosomally integrated sequences.

Occurrence in Prokaryotes

Plasmids

Plasmids are small, circular, double-stranded DNA molecules that exist independently of the bacterial , typically ranging in size from a few kilobases to several hundred kilobases. These extrachromosomal elements are self-replicating and maintain multiple copies within a single bacterial cell, enabling rapid dissemination of genetic information. In prokaryotes, plasmids are ubiquitous across bacterial and archaeal species, serving as key vehicles for that drives microbial and adaptation. Plasmid replication occurs autonomously through specific origins of replication, distinct from those of the host chromosome. For instance, the ColE1 origin, commonly found in plasmids of Escherichia coli, initiates replication via RNA priming and proceeds in a theta mode, producing bidirectional replication forks. Copy number control is achieved through regulatory mechanisms, including antisense RNA inhibition and partitioning systems involving proteins such as ParA and ParB, which actively segregate plasmids to daughter cells during division to prevent loss. Alternative replication modes include rolling-circle replication, seen in smaller plasmids like pC194 in Gram-positive bacteria, where a single-strand displacement mechanism generates linear intermediates that are later circularized. Plasmids confer diverse functional advantages to their bacterial hosts, often encoding traits that enhance survival in challenging environments. Resistance (R) plasmids carry genes for antibiotic resistance, such as those encoding beta-lactamases or efflux pumps, allowing to evade agents. The F () plasmid in E. coli enables conjugation, facilitating the transfer of genetic material between cells and often including factors that promote pathogenicity. Similarly, the Ti (tumor-inducing) plasmid in encodes enzymes for metabolism and genes that integrate into plant genomes, enabling symbiotic or pathogenic interactions with host plants. Plasmid diversity is reflected in their mobility and transfer capabilities, broadly classifying them as conjugative or non-conjugative. Conjugative plasmids, like the , possess a complete set of transfer (tra) genes that form a type IV secretion system for direct cell-to-cell DNA transfer via conjugation. Non-conjugative plasmids lack these tra genes but can be mobilized if co-resident with a conjugative plasmid, relying on shared origins of transfer (oriT) for hitchhiking during conjugation events. This mobility underpins plasmids' essential role in , allowing the spread of adaptive traits across bacterial populations without vertical inheritance.

Bacteriophage DNA

In the lysogenic cycle of temperate bacteriophages, the viral genome can exist transiently as an extrachromosomal circular DNA molecule within the prokaryotic host before potential integration into the bacterial chromosome. For bacteriophage lambda, which infects Escherichia coli, the linear double-stranded DNA genome of approximately 48.5 kb enters the host cell and rapidly circularizes via its cohesive ends, forming a covalently closed circular plasmid-like structure. This extrachromosomal form serves as the substrate for the lysogeny decision, where the phage either establishes dormancy or proceeds to the lytic cycle. The cI repressor protein, encoded by the cI gene, plays a central role in maintaining repression of lytic genes during this phase, binding to operator sites on the circular DNA to prevent expression of early lytic promoters. Unlike , which typically integrates its genome to complete lysogeny, P1 maintains its extrachromosomal state as a stable, low-copy-number throughout the lysogenic phase, with a of about 94 . P1 replication is tightly coupled to the host , occurring once per division to ensure equitable partitioning to daughter cells, facilitated by the and ParB proteins that act analogously to chromosomal segregation systems. Repression in P1 lysogens is similarly enforced by a cI-like that inhibits lytic , contributing to the phage's decision between lysogeny and upon infection, often influenced by host multiplicity of infection. Lysogenic bacteriophages confer significant biological advantages to their prokaryotic hosts through extrachromosomal maintenance. In lysogens, the cI-mediated repression provides immunity against by homologous phages, as incoming DNA cannot initiate lytic replication due to the established . Similarly, P1 plasmids impart exclusion, enhancing host survival in phage-rich environments. Beyond defense, some lysogenic phages carry accessory genes that alter host physiology; for instance, the beta-phage in encodes the tox gene for , expressed only under specific iron-limiting conditions in the lysogen, thereby converting non-pathogenic strains to toxigenic ones. The extrachromosomal phage DNA can transition to the via induction, often triggered by host DNA damage that activates the response, leading to inactivation, genome excision (if previously integrated), and subsequent virion production. In P1, this process mobilizes the for rolling-circle replication and packaging into new particles, highlighting the between and in prokaryotic systems.

Occurrence in Eukaryotes

Organellar DNA

Organellar DNA refers to the genetic material found in eukaryotic organelles, primarily mitochondria and chloroplasts, which exist as extrachromosomal elements semi-autonomous from the nuclear genome. (mtDNA) in humans is a circular, double-stranded approximately 16,569 base pairs in length. It encodes 13 proteins essential for , along with 2 ribosomal RNAs (rRNAs) and 22 transfer RNAs (tRNAs). This compact genome is maternally inherited, with paternal mtDNA typically eliminated during fertilization. Chloroplast DNA (cpDNA), present in photosynthetic eukaryotes, consists of larger circular molecules ranging from 120 to 160 kilobase pairs. These genomes encode around 100-130 genes, many involved in , including components of the , ribosomal proteins, rRNAs, and tRNAs. Unlike the strictly maternal of mtDNA, cpDNA exhibits biparental in certain plant species, such as some angiosperms, though maternal inheritance predominates in most cases. Both mtDNA and cpDNA maintain autonomy through dedicated replication and transcription machineries. For mtDNA, replication is mediated by , the sole replicase in mitochondria, which operates independently of replication processes. Transcription of mtDNA occurs within the using nuclear-encoded factors imported into mitochondria, but the process is decoupled from transcription regulation. Similarly, cpDNA replication employs -specific polymerases and proteins, often nuclear-encoded but functioning locally in the . Transcription in chloroplasts involves both plastid-encoded (PEP) and nuclear-encoded (NEP), enabling independent . The evolutionary origins of organellar DNA trace back to endosymbiotic events, with mtDNA deriving from an alpha-proteobacterium and cpDNA from a cyanobacterium. These genomes retain prokaryotic-like features, including their circular and, in the case of cpDNA, a general absence of introns in many protein-coding genes. Variants such as nuclear mitochondrial DNA segments (NUMTs) represent transferred mtDNA fragments integrated into the nuclear genome, but true extrachromosomal mtDNA persists as the functional organellar form.

Nuclear Extrachromosomal DNA

Nuclear extrachromosomal DNA (ecDNA) in eukaryotes primarily consists of non-viral circular forms known as extrachromosomal circular DNAs (eccDNAs), which range in size from approximately 100 base pairs to 1 megabase and arise through mechanisms such as gene excision from chromosomal DNA. These eccDNAs are double-stranded, closed-circle molecules that exist independently of the linear chromosomes within the nucleus. Double minutes (DMs) are small, acentric circular chromatin bodies that occur as extrachromosomal elements, particularly as precursors in cancer development, where they amplify oncogenes. EccDNAs are prevalent in specific eukaryotic cell types, including post-mitotic neurons and immune cells, where they contribute to cellular heterogeneity. In neurons, eccDNAs are abundant in both healthy and aged tissues, with tens of thousands detected in brain tissue samples from models. Their levels increase during aging, accumulating in senescent cells and promoting age-related phenotypes in and mammals. Similarly, eccDNA upregulation occurs under cellular conditions, such as or , serving as signaling molecules in immune responses. In immune cells, eccDNAs enriched with repetitive sequences activate innate immunity pathways during . Functionally, nuclear eccDNAs act as "gene parking" structures, enabling rapid amplification of gene copy numbers to facilitate quick expression of adaptive without altering the chromosomal . This allows for accelerated cellular responses, such as in evolutionary or stress survival, by increasing or essential gene dosage transiently. In B-cells, eccDNAs play a specific role in , where they are generated during heavy-chain recombination to support antibody diversification. Regulation of nuclear eccDNAs involves dynamic interactions with nuclear structures, including association with nuclear pore complexes (NPCs) via nuclear actin filaments, which facilitate their export or positioning for degradation. Epigenetic marks on eccDNAs differ from those on chromosomal DNA, often exhibiting reduced histone association and variable modifications like altered DNA methylation or histone acetylation patterns, which influence their stability and transcriptional activity. These distinct epigenetic profiles allow eccDNAs to evade typical chromosomal silencing mechanisms. A notable non-cancer example of nuclear eccDNA occurs during Drosophila development, where eccDNAs form multimers of tandemly repeated genes, including those involved in chorion production for eggshell formation, supporting developmentally timed gene amplification.

Viral Extrachromosomal DNA

Episomal Forms

Episomal forms of viral extrachromosomal DNA refer to covalently closed circular genomes of certain DNA viruses that persist in the nucleus of eukaryotic host cells without integrating into the host chromosomes. These episomes, derived from viruses such as Epstein-Barr virus (EBV), human papillomavirus (HPV), and simian virus 40 (SV40), maintain latency by replicating in synchrony with the host cell cycle using hijacked cellular machinery. Replication of these episomes relies on specific viral origins of replication (ori) and associated proteins that recruit host replication factors. In EBV, the oriP element consists of the family of repeats (FR) and dyad symmetry (DS) regions; EBNA-1 binds to DS to facilitate replication once per , mimicking chromosomal origins. Similarly, HPV episomes use the upstream regulatory region as an ori, where the E2 protein binds multiple sites to recruit the , initiating bidirectional replication dependent on host polymerases. For SV40, the viral large T antigen binds the core ori to unwind DNA and assemble the with host proteins like RPA and . During , copy numbers remain low, typically 1-50 episomes per cell for EBV and HPV in infected tissues, ensuring stable persistence without triggering lytic cycles; in contrast, lytic replication can amplify copies to hundreds or thousands to support virion production. Representative examples illustrate episomal maintenance in distinct contexts. HPV episomes persist at low copy numbers in basal of , driving epithelial proliferation through and E7 oncoproteins while replicating with host DNA. EBV episomes are maintained in B lymphocytes during latent infection, with EBNA-1 ensuring segregation; this form underlies conditions like and certain lymphomas. SV40 episomes, often studied in transformed rodent or human cells, achieve higher copy numbers (50-1000 per cell) via large T and small t antigens, contributing to cellular immortalization in experimental models. Stability of these episomes during host cell division is achieved through partitioning mechanisms where viral proteins tether the circular DNA to host chromosomes, emulating centromeric functions. EBNA-1 in EBV links to mitotic chromosomes via interactions with host factors like hEBP2, ensuring equitable distribution to daughter cells. In HPV, E2 mediates attachment to host chromatin through BRD4 binding, promoting faithful segregation in dividing . SV40 employs large T antigen for similar host interactions, maintaining episomal integrity in non-permissive human cells during persistent infection. These strategies allow long-term viral persistence without genomic .

Host Cell Interactions

Host cells recognize viral DNA, including that from extrachromosomal , through receptors. For nuclear-replicating viruses like EBV and HPV, nuclear sensors such as IFI16 detect viral genomes in the , while the cGAS-STING pathway senses cytosolic double-stranded DNA that may arise from or episome leakage during . Upon binding to foreign DNA, cyclic GMP-AMP synthase (cGAS) produces the second messenger 2'3'-cGAMP, activating (STING) to trigger type I production and release, thereby initiating innate immune defense against viruses like herpesviruses. This recognition mechanism is crucial for distinguishing self from non-self DNA, with cGAS preferentially sensing longer DNA fragments (>500–1,000 base pairs) typical of viral DNA in the . Viruses counteract this detection through immune evasion strategies, often employing viral proteins to suppress interferon responses and NF-κB signaling. For instance, Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) activates both canonical and non-canonical NF-κB pathways via its C-terminal activating regions (CTAR1 and CTAR2), recruiting TRAF adapters to induce anti-apoptotic and immunomodulatory cytokines like IL-10 and IL-6, which dampen type I interferon signaling and promote B-cell survival. LMP1 further inhibits retinoic acid-inducible gene I (RIG-I) and Toll-like receptor 9 (TLR9) expression, reducing innate sensing of viral DNA and facilitating persistent infection. Viral ecDNA interacts with host cellular machinery to ensure stability, such as tethering episomes to mitotic chromosomes for faithful segregation during . In EBV, the Epstein-Barr nuclear antigen 1 (EBNA1) binds to specific AT-rich docking sites on chromosomes marked by , using its arginine-glycine repeats and AT-hook domains to mediate homotypic interactions and , often repressing nearby genes like neuronal factors (e.g., NRXN1). Similarly, human (HCMV) genomes associate with chromosome peripheries in infected cells, supported by immediate-early protein 1 (IE1), enabling genome retention without integration. If undetected, these interactions promote viral persistence; however, failure to evade can induce arrest or via activation and DNA damage responses. A key example is human papillomavirus (HPV), where E6 and E7 oncoproteins degrade the tumor suppressor through E6AP-mediated ubiquitination, disrupting and to allow episomal maintenance in epithelial cells. This evasion enables establishment by inhibiting host clearance mechanisms like ATM-CHK2 signaling, contrasting with effective immune clearance in immunocompetent hosts that limits viral spread. Unchecked persistence contributes to oncogenesis, as seen in EBV-driven lymphomas and HPV-associated cervical cancers, where dysregulated programs induce genomic instability and proliferation without triggering full immune elimination.

Inheritance and Dynamics

Segregation and Stability

Extrachromosomal DNA (ecDNA) exhibits distinct segregation mechanisms depending on its context within prokaryotic or eukaryotic cells. In nuclear ecDNA, commonly observed in cancer cells, occurs randomly during due to the absence of centromeres, resulting in unequal partitioning to daughter cells. This contrasts with chromosomal inheritance and promotes rapid heterogeneity. In contrast, bacterial plasmids employ active segregation systems, such as the ParMRC apparatus, where ParM actin-like filaments push plasmid copies toward opposite cell poles, ensuring equitable division and high fidelity inheritance. Stability of ecDNA is maintained through compensatory mechanisms that offset segregation losses. High copy number amplification allows nuclear ecDNA to persist despite random partitioning, as elevated copies increase the probability of retention in both daughter cells; for instance, oncogene-bearing ecDNA in tumors can amplify under selective pressure to sustain function. Additionally, some ecDNA elements tether to chromosomes, enhancing co- and reducing missegregation into the . For (), a genetic during reduces effective copy number to a few dozen, enabling near-Mendelian and rapid resolution across generations. Segregation rates vary significantly, with nuclear eccDNA experiencing 10-50% loss per in the absence of selection, leading to progressive dilution unless counteracted by replication. These rates are influenced by cell type—plasmids remain stable in through par-mediated partitioning, while ecDNA shows high variability in tumor cells—and environmental factors, such as stress or , which accelerate loss by suppressing replication. modeling, using algorithms like Gillespie simulations, demonstrates how random generates broad copy number distributions and intratumoral heterogeneity, facilitating adaptive evolution in dynamic environments.

Evolutionary Roles

Extrachromosomal DNA (ecDNA), including plasmids and DNA, plays a pivotal role in (HGT), enabling rapid dissemination of adaptive traits across bacterial populations. Plasmids, as self-replicating extrachromosomal elements, facilitate conjugation-mediated transfer of genes conferring antibiotic resistance, accelerating evolutionary adaptation in response to selective pressures like antimicrobial exposure. further enhance this process through , packaging and delivering bacterial DNA, including resistance genes, between hosts, which has been instrumental in the global spread of multidrug-resistant strains. Phage-plasmids, hybrid elements combining features of both, promote recombination and the emergence of novel resistance cassettes, underscoring ecDNA's contribution to microbial evolution beyond vertical inheritance. In unicellular organisms, ecDNA drives by serving as a for duplications, deletions, and rearrangements, allowing swift adjustments to environmental challenges. These circular molecules can amplify copy numbers without disrupting chromosomal integrity, providing a for rapid in fluctuating conditions, such as or . For instance, eccDNA formation enables unicellular eukaryotes and prokaryotes to generate genetic variants at higher rates than chromosomal mutations, fostering population-level diversity and survival advantages. Among eukaryotes, mitochondrial DNA (mtDNA), a vestigial extrachromosomal element derived from an ancient bacterial endosymbiont, harbors mutations that propel metabolic evolution by altering energy production pathways. These mutations, often heteroplasmic, influence cellular respiration efficiency and have shaped eukaryotic diversification across lineages. Nuclear ecDNA, meanwhile, contributes to immune system evolution; byproducts of V(D)J recombination in lymphocytes form circular DNAs that diversify antigen receptor genes, enhancing adaptive immunity and host defense variability. In cancer contexts, recent 2025 analyses reveal that ecDNA enables non-Mendelian inheritance in tumors, allowing oncogene amplification to propagate asymmetrically during cell division and accelerate tumor evolution under therapeutic selection, including through RNA-mediated tethering and selective degradation. On a broader scale, ecDNA has influenced through the retention of endosymbiont-derived genomes, as seen in the endosymbiotic origin of mitochondria, where partial retention in mtDNA facilitated eukaryotic cellular complexity and divergent evolutionary trajectories. Recent 2024 reviews highlight ecDNA's role in microbial , where plasmids and phage-derived elements mediate community dynamics, , and niche adaptation in diverse environments like and aquatic systems.

Detection Methods

Traditional Approaches

Early methods for detecting and characterizing extrachromosomal DNA (ecDNA) relied on and basic biochemical separation techniques, which laid the foundation for understanding its structure and presence in eukaryotic cells. In the 1960s, electron emerged as a pivotal tool for visualizing circular DNA configurations, particularly in organelles. Margit M. K. Nass and Sylvan Nass first observed intramitochondrial fibers with DNA-like staining properties in mouse liver cells using electron and specific fixation techniques, providing initial evidence of extrachromosomal DNA outside the . Subsequent studies confirmed the circular nature of through electron micrographs of extracted molecules, revealing theta structures indicative of replication and closed-loop forms distinct from linear chromosomal DNA. These observations extended to in plants, where similar circular forms were identified via electron in the late 1960s. In cancer during the 1970s, light microscopy identified double minutes—small, paired extrachromosomal chromatin bodies lacking centromeres—in tumor metaphase spreads, marking early recognition of nuclear ecDNA in mammalian cells. (FISH), an advancement building on these cytogenetic approaches, began to be applied in the late 1970s and early 1980s to map specific sequences on double minutes, confirming their extrachromosomal origin and gene content in cancer lines like COLO 320. However, FISH at the time was limited by probe resolution and required fixed preparations, often complementing traditional staining methods. Density centrifugation provided a key biochemical approach for isolating ecDNA based on buoyant density differences from DNA. In the , cesium chloride (CsCl) density centrifugation separated , enabling purification from total cellular extracts. This method, often combined with to distinguish supercoiled circular forms, was routinely used to isolate in plants, isolating closed circular molecules of 100-150 kb. Such techniques confirmed the organellar localization and purity of ecDNA fractions for downstream analyses. Southern blotting, developed in the mid-1970s, allowed detection of non-integrated viral ecDNA by hybridizing restriction-digested DNA with radiolabeled probes. Edwin Southern's 1975 method transferred electrophoresed DNA fragments to membranes, enabling specific identification of episomal viral genomes like Epstein-Barr virus in latently infected cells, where supercoiled forms produced characteristic band patterns distinct from integrated sequences. This approach was widely adopted for quantifying extrachromosomal viral DNA copies in host cells by the late 1970s. Pulsed-field gel electrophoresis (PFGE), introduced in the 1980s, facilitated separation of large ecDNA molecules from chromosomal DNA. By alternating directions, PFGE resolved DNA fragments up to several megabases, allowing isolation of intact extrachromosomal circles or double minutes from yeast artificial chromosomes or cancer cell lines without shearing. This technique proved essential for characterizing large nuclear ecDNA in mammalian cells, distinguishing them from linear chromosomes based on migration patterns. Despite these advances, traditional approaches had notable limitations, including low sensitivity for detecting low-copy-number ecDNA amid high nuclear DNA background and challenges in sequencing small circular molecules due to the lack of high-throughput tools. often required labor-intensive and provided only structural insights without sequence information, while separation methods like density gradients and PFGE were prone to and inefficient for trace amounts. Southern blotting, though specific, depended on prior knowledge of sequences and could not resolve structural variants in ecDNA.

Contemporary Techniques

Next-generation sequencing (NGS) has revolutionized ecDNA detection through read-depth analysis, which quantifies copy number variations by comparing sequencing coverage across genomic regions to identify focal amplifications indicative of extrachromosomal elements. This approach is particularly effective in cancer samples, where ecDNA often drives overexpression via high copy numbers, as demonstrated in cohorts where read-depth thresholds above 10-fold amplification signal ecDNA presence. To specifically enrich circular DNA prior to NGS, Circle-seq utilizes digestion to degrade linear DNA while preserving circles, followed by library preparation and sequencing; this method achieves over 100-fold enrichment for eccDNA ranging from 100 bp to megabases, uncovering genome-wide circular structures in tumors. An enhanced 2025 protocol for Circle-seq addresses biases in enrichment efficiency, improving detection sensitivity for low-abundance ecDNA in heterogeneous samples. Long-read sequencing platforms, including PacBio HiFi and , enable resolution of complete ecDNA structures by producing reads exceeding 10 kb, which span junction breakpoints and repetitive regions inaccessible to short-read NGS. These technologies identify chimeric junctions formed during ecDNA biogenesis, such as head-to-tail concatemers in oncogene-amplified circles. In 2024 protocols applied to advanced cancer cohorts, long-read sequencing resolved ecDNA harboring viral integrations and complex rearrangements in over 80% of predicted cases, with tools like assembling full ecDNA contigs from raw reads using graph-based algorithms that model discordant alignments. This has facilitated annotation of ecDNA in patient-derived models, revealing structural heterogeneity not captured by short reads. Microscopy advancements, particularly super-resolution techniques like STED and , provide nanometer-scale visualization of ecDNA localization and dynamics within the , distinguishing extrachromosomal hubs from chromosomal integrations. HaloTag labeling enables live-cell tracking of ecDNA partitioning during , showing asymmetric inheritance in cancer cells. Complementing this, assays accessibility on ecDNA, revealing elevated open regions in amplified oncogenes like , with peak signals 2-5 times higher than in linear DNA; this method detected pre-amplification ecDNA in therapy-resistant tumors, predicting resistance emergence. Bioinformatics pipelines exploit discordant read mapping in NGS data to pinpoint ecDNA, where reads with unexpected orientations or spanning non-adjacent genomic loci indicate circular junctions. Tools such as ECCsplorer process BAM files to filter split and paired-end discordant reads, achieving >95% specificity in identifying ecDNA under 1 Mb across diverse tissues. Similarly, ecc_finder models read-pair distances to cluster potential circles, validated on simulated datasets with 90% recall for low-coverage samples. models further enhance prediction from whole-genome sequencing by training on features like read-depth variance and density; for instance, a 2024 classifier detects ecDNA in whole-exome data with 85% accuracy, prioritizing high-impact amplifications in pan-cancer analyses. Innovations in 2025 include single-cell ecDNA profiling via scATAC-seq, which captures accessibility patterns at cellular resolution to infer ecDNA in heterogeneous populations, identifying amplified foci in 20-30% of tumor cells missed by bulk methods. The ATACAmp algorithm analyzes scATAC-seq peaks for copy number anomalies, enabling ecDNA/HSR discrimination in single nuclei. Integration with CRISPR-Cas9 supports functional validation by targeted cleavage or enrichment of specific ecDNA, as in CRISPR-CATCH protocols that isolate oncogene-bearing circles for downstream phenotyping, confirming their role in with >50% reduction upon disruption.

Biological Significance

Functions in Normal Cells

In normal cells, extrachromosomal DNA (ecDNA) serves as a transient amplifier for during developmental processes, particularly in oocytes where it facilitates rapid production of essential proteins. For instance, in amphibian oocytes such as those of Xenopus laevis, (rDNA) is amplified extrachromosomally to generate multiple copies of the genes encoding , supporting the high demand for synthesis during . This amplification occurs through rolling-circle replication, producing free extrachromosomal circles that are not integrated into the nuclear genome, thereby enabling efficient, stage-specific without altering chromosomal structure. In the immune system, ecDNA arises as a byproduct of V(D)J recombination, a physiological process that assembles diverse antigen receptor genes in developing lymphocytes. During this recombination, the RAG1 and RAG2 proteins cleave DNA at recombination signal sequences, excising intervening segments that form stable extrachromosomal signal joint circles. These circles, such as T-cell receptor excision circles (TRECs) in T cells and kappa-deleting recombination excision circles (KRECs) in B cells, persist transiently without integrating into the genome, contributing to immune repertoire diversity by allowing precise joining of variable (V), diversity (D), and joining (J) segments while maintaining genomic stability. ecDNA also supports in prokaryotes through plasmids, which are autonomously replicating extrachromosomal elements that confer environmental responsiveness. In like species, plasmids carry genes for metabolic adjustments and resistance, enabling rapid adaptation to host intracellular niches via and coevolution with chromosomal elements. Similarly, (mtDNA), a circular extrachromosomal , maintains in eukaryotic cells by encoding components of the system, with its copy number regulated to match cellular metabolic demands through and dynamics. Tissue-specific variations in ecDNA abundance reflect physiological roles, with higher levels observed in dynamic tissues like the compared to stable somatic cells. In murine brain tissues, such as and , eccDNA correlates with open regions marked by H3K27ac and H3K4me1, potentially supporting neuronal by influencing near immediate-early genes involved in synaptic remodeling. In contrast, tissues like exhibit lower eccDNA levels, consistent with their post-mitotic stability. In plants, (cpDNA), an extrachromosomal genome, sustains photosynthetic efficiency by encoding proteins for the and , optimizing light energy conversion in leaf mesophyll cells. Certain viral episomes maintain persistent infections in normal host cells without disrupting physiology, acting as stable extrachromosomal replicons. For example, (HSV-1) persists as low-copy episomes in sensory neurons, regulated by latency-associated transcripts to ensure lifelong carriage with minimal host impact until reactivation triggers. This episomal form allows the virus to evade immune clearance while integrating into the host's cellular .

Implications in Disease

Extrachromosomal DNA (ecDNA), including mitochondrial DNA (mtDNA) deletions and nuclear eccDNA, plays a significant role in neurodegenerative diseases such as Parkinson's disease. In Parkinson's, the 4977-bp common mtDNA deletion, spanning from nucleotide 8470 to 13447, accumulates in substantia nigra neurons and is associated with mitochondrial dysfunction and dopaminergic cell loss. This deletion disrupts oxidative phosphorylation by affecting genes like MT-ATP8 and MT-ND5, contributing to energy deficits observed in affected brain regions. Additionally, nuclear eccDNA accumulates in aging brains, potentially exacerbating genomic instability and neuronal vulnerability, as evidenced by increased eccDNA profiles in aged mouse brain structures compared to young ones. In infectious diseases, viral episomes serve as extrachromosomal elements that promote persistence. Epstein-Barr virus (EBV) maintains latency in B cells during by circularizing into episomal form, tethered to host via EBNA1, allowing lifelong infection without integration. Similarly, unintegrated HIV-1 DNA contributes to viral latency by persisting as extrachromosomal forms in infected cells, enabling low-level and evasion of immune detection, which sustains chronic infection. These episomal structures facilitate reactivation and complicate eradication efforts in latency reservoirs. Genetic disorders involving ecDNA often arise from nuclear-mitochondrial transfers, such as numts (nuclear mitochondrial DNA segments), which integrate mtDNA fragments into the nuclear genome as nonfunctional pseudogenes. Numts can introduce sequencing artifacts or mimic mtDNA variants, complicating diagnostics in mitochondrial disorders, and their accumulation is linked to diseases through erroneous or genomic instability. In mitochondrial diseases like , caused by mtDNA point mutations such as m.3243A>G in MT-TL1, extrachromosomal mtDNA dynamics—resembling plasmid-like circular elements—contribute to shifts and impaired tRNA function, leading to encephalomyopathy and . EcDNA also influences autoimmunity by acting as autoantigens that trigger aberrant immune responses. Circulating eccDNA, derived from apoptotic cells, acts as a novel autoantigen in systemic lupus erythematosus (SLE), where plasma eccDNA profiles differ from healthy controls and correlate with immunological markers such as complement C3 and . In chronic infections, bacterial plasmids persist as extrachromosomal elements, conferring antibiotic resistance and virulence factors that sustain reservoirs.

Role in Cancer

Oncogene Amplification

Extrachromosomal DNA (ecDNA) serves as a primary mechanism for oncogene amplification in cancer, enabling high copy numbers of key driver genes that drive tumorigenesis through overexpression. Unlike chromosomal amplifications, ecDNA can exist in tens to hundreds of copies per cell, such as for oncogenes like MYC and EGFR, resulting in substantially elevated transcript levels compared to equivalent linear DNA amplifications. This amplification occurs via focal excision and circularization of genomic regions containing oncogenes, often facilitated by chromothripsis—a catastrophic shattering of chromosomes within micronuclei during mitosis—that generates the initial DNA fragments for ecDNA formation. Under selective pressure in the tumor microenvironment, clones harboring these amplified ecDNAs proliferate preferentially, accelerating tumor progression and heterogeneity. In specific cancers, ecDNA-mediated oncogene amplification is particularly prevalent and impactful. For instance, in , approximately 90% of MYCN amplifications occur on ecDNA, contributing to aggressive disease phenotypes and poor by hijacking distal enhancers that boost MYCN expression. Similarly, in , the EGFRvIII variant—a constitutively active —is frequently amplified on ecDNA, where it associates with active enhancers to drive rapid tumor evolution and therapy resistance. These examples highlight how ecDNA enables dynamic, high-level oncogene dosage that outpaces chromosomal mechanisms, with ecDNA detected across 10–60% of cases in diverse tumor types, including up to 49% in glioblastomas and 55% in liposarcomas. The circular structure of ecDNA confers advantages over linear amplifications, including evasion of TP53-dependent degradation pathways that target unstable linear DNA, thereby promoting persistence and accumulation in cancer cells. Additionally, ecDNA's lack of centromeres leads to random, heterogeneous segregation during , generating subclonal variation in oncogene copy number that fuels rapid tumor and . Recent findings further reveal that ecDNA integrates into nuclear condensates via association with the MED1 transcription coactivator, forming hubs that reorganize and enhance oncogenic transcription in a cancer-type-specific manner. This condensate-mediated boosting of underscores ecDNA's role in sustaining high output.

Therapeutic Resistance

Extrachromosomal DNA (ecDNA) facilitates rapid evolutionary adaptation in cancer cells, enabling quick shifts in expression under therapeutic pressure. This non-chromosomal allows for asymmetric segregation during , promoting stochastic changes in that accelerate the selection of resistant clones. For instance, in response to targeted therapies, ecDNA can drive the emergence of alternative oncogenic pathways, enhancing tumor survival and progression. The mosaic distribution of ecDNA within tumors generates significant intratumoral heterogeneity, fostering subpopulations with varying sensitivities to . This uneven partitioning leads to phenotypic , where cells with higher ecDNA copy numbers exhibit enhanced and survival advantages during . A 2024 pan-cancer analysis revealed that ecDNA presence correlates with poor , including shorter overall survival (hazard ratio 1.44) and increased likelihood of advanced disease stages, underscoring its role in driving resistant subpopulations. In MYCN-amplified neuroblastoma, ecDNA-driven oncogene dosage heterogeneity promotes rapid adaptation to , with senescent cells harboring lower ecDNA copies contributing to . Key mechanisms of ecDNA-mediated resistance include the amplification of drug efflux genes, such as MDR1, which encodes to expel chemotherapeutic agents like and . In human epidermal carcinoma cells, MDR1 amplification on ecDNA increases gene copy number and expression, directly enhancing multidrug resistance through ATP-dependent efflux pumps. Additionally, ecDNA carrying enables bypass signaling pathways by upregulating activity, allowing cells to evade therapy-induced dependencies; for example, in pancreatic ductal , ecDNA confers independence from stromal WNT signals, promoting plasticity and survival under stress. Clinically, ecDNA is strongly associated with relapse in various cancers, including ovarian and breast tumors, where its retention in recurrent lesions outpaces chromosomal amplifications. In high-grade serous ovarian cancer, ecDNA contributes to clonal evolution and therapeutic failure, while in breast cancer, it drives oncogene amplification linked to distant metastasis in up to 30% of early-stage cases. A 2025 preprint highlights ecDNA's role in fast-tracking evolutionary adaptability at the population level, making tumors resilient to interventions and prone to recurrence.01620-X/fulltext) A major challenge in targeting ecDNA arises from its dynamic nature, where copy numbers may decline post-treatment due to selective pressures but re-emerge from residual heterogeneous cells during . This transient loss, observed in models of on MDR1-amplified ecDNA, complicates eradication efforts, as surviving subpopulations can regenerate ecDNA under renewed stress, perpetuating resistance.

References

  1. [1]
    Extrachromosomal circular DNA: biogenesis, structure, functions ...
    Oct 2, 2022 · Extrachromosomal circular DNA (eccDNA) is a collective name for the circular, double-stranded molecules in the nuclei, which is derived from but independent of ...
  2. [2]
    Origins and impact of extrachromosomal DNA - Nature
    Nov 6, 2024 · Extrachromosomal DNA (ecDNA) is a major contributor to treatment resistance and poor outcome for patients with cancer.
  3. [3]
    Extrachromosomal DNA (ecDNA) in cancer: mechanisms, functions ...
    Jun 28, 2023 · Extrachromosomal DNA (ecDNA) is circular DNA that plays an important role in the development and heterogeneity of cancer.
  4. [4]
  5. [5]
    Extrachromosomal circular DNA: Current status and future prospects
    Oct 18, 2022 · ... defining characteristics should be ... extrachromosomal DNA elements and transcriptionally active extrachromosomal DNA hubs in cancer.
  6. [6]
    History of chloroplast genomics | Photosynthesis Research
    The presence of chloroplast DNA was established in 1963. With the development of recombinant DNA technologies, chloroplast DNA was selected as one of the ...
  7. [7]
    Life of double minutes: generation, maintenance, and elimination - NIH
    Apr 30, 2022 · Double minutes harbor oncogenes or drug resistance genes that contribute to tumor aggressiveness through copy number amplification.
  8. [8]
    Modern biology of extrachromosomal DNA: A decade-long voyage ...
    acentric, circular DNA molecules ranging from 50 ...
  9. [9]
  10. [10]
    Overlooked: Extrachromosomal DNA and Their Possible Impact on ...
    Apr 27, 2018 · This review raises critical questions about the reliance on so-called genome sequencing data of whole DNA, in particular of human genomes.Missing: definition | Show results with:definition
  11. [11]
    Targeted profiling of human extrachromosomal DNA by CRISPR ...
    Oct 17, 2022 · We demonstrate strong enrichment of ecDNA molecules containing EGFR, FGFR2 and MYC from human cancer cells and NRAS ecDNA from human metastatic melanoma.Missing: properties | Show results with:properties
  12. [12]
    Extrachromosomal DNA (ecDNA): an origin of tumor heterogeneity ...
    Nov 10, 2022 · Second, the circular topology of ecDNA is characterized by highly accessible chromatin (see Box 2), a paucity of repressive histone marks with ...
  13. [13]
    Enhancing transcription–replication conflict targets ecDNA ... - Nature
    Nov 6, 2024 · ecDNA exhibits open chromatin and is marked by active histone modifications such as H3K27ac and H3K4me3 (refs.), raising the possibility of a ...
  14. [14]
    Scaling laws of bacterial and archaeal plasmids - Nature
    Jul 2, 2025 · Plasmids are extrachromosomal DNA elements, found ubiquitously across bacteria and archaea, that mediate the flow of genes within and across ...
  15. [15]
    Replication and Control of Circular Bacterial Plasmids - PMC
    There are three general replication mechanisms for circular plasmids, namely, theta type, strand displacement, and rolling circle (RC). Historical development ...
  16. [16]
    Pathways for horizontal gene transfer in bacteria revealed by a ...
    Jul 17, 2020 · Plasmids can mediate horizontal gene transfer of antibiotic resistance, virulence genes, and other adaptive factors across bacterial populations.
  17. [17]
    Coordinating plasmid partition with bacterial chromosome segregation
    May 9, 2024 · Plasmid partition is a transport process, in that interactions between ParA and ParB drive the movement of plasmid DNA cargo via ParB/parS partition complexes.Missing: ColE1 | Show results with:ColE1
  18. [18]
    A Brief History of Plasmids - PMC - PubMed Central
    The foundations of plasmid biology were built largely on careful genetic analysis of the properties of the F plasmid, F-prime derivatives, R plasmids, and Col ...
  19. [19]
    Plasmid Transfer by Conjugation in Gram-Negative Bacteria
    Here, we review the key steps of plasmid transfer by conjugation in Gram-negative bacteria, by following the life cycle of the F factor during its transfer.Missing: Ti | Show results with:Ti
  20. [20]
    Agrobacterium-mediated plant transformation: biology and ...
    This article aims to provide a comprehensive review of the biology and the applications of Agrobacterium-mediated plant transformation
  21. [21]
    Genomics of microbial plasmids: classification and identification ...
    Mar 30, 2015 · Conjugative transfer is another important mechanism by which plasmids spread DNA among different bacteria. Self-transmissible plasmids in Gram- ...
  22. [22]
    Origin-of-transfer sequences facilitate mobilisation of non ...
    Conjugative plasmids can facilitate the transfer of non-conjugative plasmids present in the same cell; a process known as mobilisation. It is generally accepted ...Results · Orit Sequences Carry... · Smpo Acts As An Orit...
  23. [23]
    The protein interaction map of bacteriophage lambda
    Sep 26, 2011 · Two of the best-characterized aspects of lambda biology are the genetic switch that determines whether a phage reproduces and lyses the cell or ...
  24. [24]
    Bacteriophage lambda: early pioneer and still relevant - PMC
    In this review we give some relevant early history and describe recent developments in understanding the molecular biology of lambda's life cycle.
  25. [25]
    ISOLATION OF THE λ PHAGE REPRESSOR - PNAS
    Here we reveal maps of bodily sensations associated with different emotions using a unique topographical self-report method.
  26. [26]
    Genome of Bacteriophage P1 | Journal of Bacteriology
    As a prophage, P1 is a stable plasmid maintained at about one copy per bacterial chromosome. Several P1 genes scattered throughout the genome are expressed in ...
  27. [27]
    Maintenance of bacteriophage P1 plasmid - PMC - NIH
    Three mutants of bacteriophage P1 affected in their ability to maintain the lysogenic state stably are described here. These mutants were normal in lytic ...
  28. [28]
    Corynebacterium diphtheriae: Diphtheria Toxin, the tox Operon, and ...
    In its lysogenic state, most β-phage genes appear to be repressed, and the lysogen is immune to superinfection by homoimmune corynebacteriophages. While ...
  29. [29]
    Host responses influence on the induction of lambda prophage - NIH
    Prophage induction can be initiated either by DNA damage or by heat treatment of a temperature-sensitive repressor. These two treatments also cause a concurrent ...Introduction · Fig. 2 · Ftsh And Hflkc Are...
  30. [30]
    The little big genome: the organization of mitochondrial DNA - PMC
    The mitochondrial genome in humans contains 16,569 base pairs that code for 37 genes: 13 polypeptides, 22 tRNAs, and the small and large rRNA subunits (4).
  31. [31]
    Maternal inheritance of human mitochondrial DNA - PubMed - NIH
    The results of this study demonstrate that human mitochondrial DNA is maternally inherited. The techniques described for using peripheral blood platelets as a ...
  32. [32]
    Towards the Well-Tempered Chloroplast DNA Sequences - PMC - NIH
    Jul 2, 2021 · The chloroplast genome (cpDNA) is a double-stranded DNA molecule of about 120 kb to 160 kb in size that encodes about 100 proteins [5,6]. This ...
  33. [33]
    Chloroplast competition is controlled by lipid biosynthesis in evening ...
    Most organelle genomes are inherited from the mother (1, 2), but biparental transmission of plastids has evolved independently multiple times. Approximately 20% ...<|control11|><|separator|>
  34. [34]
    DNA polymerases in the mitochondria: A critical review of the evidence
    Since 1970, the DNA polymerase gamma (PolG) has been known to be the DNA polymerase responsible for replication and repair of mitochondrial DNA.
  35. [35]
    Nuclear transcription factors in mammalian mitochondria - PMC
    Mitochondrial DNA is packaged, handled and inherited independently of the nuclear genome, and far less is known about the regulation of mitochondrial gene ...
  36. [36]
    Chloroplast DNA Replication Is Regulated by the Redox State ... - NIH
    Chloroplasts are semiautonomous organelles that possess their own genome, which is complexed with proteins to form nucleoids and also certain machinery needed ...
  37. [37]
    Chloroplast gene expression: Recent advances and perspectives
    Transcription. Chloroplast genes are transcribed by two distinct polymerases: nucleus-encoded polymerase (NEP) and plastid-encoded polymerase (PEP). NEP is a ...
  38. [38]
    Mitochondrial Evolution - PMC - NIH
    Accordingly, the endosymbiont hypothesis—the idea that the mitochondrion evolved from a bacterial progenitor via symbiosis within an essentially eukaryotic host ...
  39. [39]
    Genomics & Chloroplast Evolution: Cyanobacteria's Role?
    Plant chloroplast originated, through endosymbiosis, from a cyanobacterium, but the genomic legacy of cyanobacterial ancestry extends far beyond the chloroplast ...
  40. [40]
    The Mighty NUMT: Mitochondrial DNA Flexing Its Code in the ... - NIH
    Apr 27, 2023 · Nuclear-mitochondrial DNA segments (NUMTs) are mitochondrial DNA (mtDNA) fragments that have been inserted into the nuclear genome.
  41. [41]
    Extrachromosomal circular DNA: biogenesis, structure, functions ...
    Oct 2, 2022 · Extrachromosomal circular DNA (eccDNA), ranging in size from tens to millions of base pairs, is independent of conventional chromosomes.Missing: seminal | Show results with:seminal
  42. [42]
    Extrachromosomal Circular DNA (eccDNA): From Chaos to Function
    Jan 5, 2022 · EccDNA is a type of double-stranded circular DNA that is derived and free from chromosomes. It has a strong heterogeneity in sequence, length, and origin.Missing: seminal nuclei
  43. [43]
    The extrachromosomal circular DNA atlas of aged and young mouse ...
    Mar 27, 2024 · On average, the number of eccDNAs per brain structure in the aged group was 20,879 (ranging from 5,662 to 75,197), while in the young group it ...
  44. [44]
    Extrachromosomal circular DNA drives dynamic genome plasticity
    The role of eccDNA extends beyond oncology into various organ systems. In neural tissues, eccDNA has been detected in both healthy neurons and brain tumors.
  45. [45]
    Extrachromosomal Circular DNA (eccDNA): From Chaos to Function
    Jan 6, 2022 · 3) Aging. EccDNA has been found accumulated in old cells and inducing aging in yeast (Hull et al., 2019). It was also amplified during aging ...Missing: prevalence | Show results with:prevalence
  46. [46]
    Extrachromosomal circular DNA drives dynamic genome plasticity
    In immune cells, eccDNA plays immunomodulatory roles, especially under stress or during apoptosis. EccDNA enriched with repetitive sequences can activate ...
  47. [47]
    Molecular mechanisms of extrachromosomal circular DNA formation
    Mar 4, 2025 · In unicellular organisms, the ability for eccDNA to rapidly modify gene copy numbers and thereby provide accelerated adaptation can provide ...
  48. [48]
    Transcription-induced formation of extrachromosomal DNA during ...
    Extrachromosomal circular DNA (eccDNA) facilitates adaptive evolution by allowing rapid and extensive gene copy number variation and is implicated in the ...
  49. [49]
    Extrachromosomal circular DNA is common in yeast - PNAS
    In humans, eccDNAs are produced during Ig heavy-chain class switching (9). In higher plant genomes, repeat-derived eccDNAs are considered intermediates in the ...
  50. [50]
    Extrachromosomal circular DNA (eccDNA): an emerging star in cancer
    Jul 26, 2022 · EccDNA is formed mainly from DNA damage repair. They then follow nuclear actin to the nuclear pore complexes (NPCs) and are eventually excluded ...
  51. [51]
    Connecting the dots: Epigenetic regulation of extrachromosomal ...
    Extrachromosomal DNA (ecDNA) amplifications can exist in both circular (i.e. extrachromosomal circular DNA; eccDNA) and linearized forms. Gene amplifications ...
  52. [52]
    an integrated resource and analysis platform for human eccDNAs
    Nov 18, 2021 · It classifies them into six sections including targeting genes, epigenetic regulations, regulatory elements, chromatin accessibility, chromatin ...
  53. [53]
    Evidence for rolling circle replication of tandem genes in Drosophila
    We showed that the eccDNA population contains circular multimers of tandemly repeated coding genes, such as histones, rDNA, Stellate and the Suppressor of ...
  54. [54]
    Control of Viral Latency by Episome Maintenance Proteins - PMC
    Oct 14, 2019 · Episome maintenance is an essential part of the life cycle of the persistent DNA tumor viruses KSHV, EBV, and HPV. Disruption of episome ...Viral Episome Maintenance... · Chromosome Tethering Of Emps · Epigenetic Programming Of...<|control11|><|separator|>
  55. [55]
  56. [56]
    The cGAS-STING Defense Pathway and Its Counteraction by Viruses
    Feb 10, 2016 · cGAS and STING have been identified as intracellular sensors that activate the interferon pathway in response to virus infection and thus mediate host defense.
  57. [57]
    Research Advances in How the cGAS-STING Pathway ... - Frontiers
    The ability of human cGAS to discriminate long dsDNA strands from shorter dsDNA may contribute to the specific sensing and recognition of the “danger DNA” of ...
  58. [58]
    Deciphering the Role of Epstein–Barr Virus Latent Membrane ...
    Overall, it seems that the goal of this LMP1 regulation is to limit its own accumulation to provide EBV-infected cells a direct escape from the host immune ...
  59. [59]
    Epigenetic specifications of host chromosome docking sites ... - Nature
    Feb 13, 2020 · EBV episome tethering to metaphase chromosomes is essential for viral genome maintenance in proliferating cells. While several mechanisms ...
  60. [60]
    Evidence for Tethering of Human Cytomegalovirus Genomes to Host ...
    Sep 30, 2020 · These viruses often depend on episome tethering to mitotic chromosomes for segregation and partitioning of their genomes to daughter cell ...
  61. [61]
    Molecular mechanisms of viral oncogenesis in humans - PMC
    Recent advances have revealed that seven known oncogenic viruses promote tumorigenesis through shared host cell targets and pathways.
  62. [62]
    The Role of HPV E6 and E7 Oncoproteins in HPV-associated ... - NIH
    Low risk and cutaneous epithelia-infecting HPV E6 proteins are unable to target the cellular p53 protein for degradation through the proteasome pathway.
  63. [63]
  64. [64]
    The evolutionary dynamics of extrachromosomal DNA in human ...
    Sep 19, 2022 · Oncogene amplification on extrachromosomal DNA (ecDNA) is a common event, driving aggressive tumor growth, drug resistance and shorter survival.
  65. [65]
    The ParMRC system: molecular mechanisms of plasmid segregation ...
    Sep 16, 2010 · The ParMRC segregation system moves plasmids to the very ends of the cell, close to the cell poles and beyond the end of the nucleoid.Missing: par | Show results with:par
  66. [66]
    Large-scale genetic analysis reveals mammalian mtDNA ... - Nature
    Jun 27, 2018 · A process known as the mtDNA bottleneck acts to increase heteroplasmy variance during development. This increase in variance allows a ...
  67. [67]
    The evolution of plasmid-carried antibiotic resistance
    May 19, 2011 · Bacterial cells are capable of transferring genes horizontally. This DNA transfer can take place in three ways, through plasmids, phages, or ...Methods And Results · Model With Non-Coding... · Discussion<|separator|>
  68. [68]
    Phage-plasmids promote recombination and emergence of ... - Nature
    Feb 20, 2024 · Phages and plasmids are regarded as distinct types of mobile genetic elements that drive bacterial evolution by horizontal gene transfer.Phage-Plasmid Gene... · Phage-Plasmids Drive Gene... · Many Plasmids And A Few...
  69. [69]
    Phage-Plasmids Spread Antibiotic Resistance Genes through ...
    Sep 26, 2022 · This study shows that phage-plasmids carry a large diversity of clinically relevant antibiotic resistance genes that they can transfer across bacteria.Phage-Plasmids Spread... · Resistance Genes Are Found... · Phage-Plasmids With...
  70. [70]
    Evolution and maintenance of mtDNA gene content across eukaryotes
    As well as driving mitochondrial evolution across eukaryotes, mtDNA mutations have important translational consequences. ... mitochondrial DNA degeneration and ...
  71. [71]
    Landscape of extrachromosomal DNA characteristics in high-grade ...
    ... DNA as a by-product of V(D)J recombination in normal tissues.,,. In the meantime, our data revealed that circular DNAs harboring intact oncogenes ...
  72. [72]
    Extrachromosomal DNA Gives Cancer a New Evolutionary Pathway
    Apr 29, 2025 · This population-based adaptability makes ecDNA-driven cancers exceptionally resilient, offering new insights into their persistence and ...Missing: Mendelian | Show results with:Mendelian
  73. [73]
    Endosymbiosis and its implications for evolutionary theory - PNAS
    This paper will discuss how metabolic innovations associated with endosymbioses can drive evolution and thus provide an explanatory account of important ...
  74. [74]
    Phage-plasmids: missed links between mobile genetic elements
    Mar 21, 2024 · Plasmids are typically extrachromosomal elements, some of which can transfer themselves and cargo genes via conjugation. Phages are bacterial ...Missing: antibiotic | Show results with:antibiotic
  75. [75]
    Specific Gene Amplification in Oocytes - Science
    Specific Gene Amplification in Oocytes: Oocyte nuclei contain extrachromosomal replicas of the genes for ribosomal RNA. ... Extrachromosomal Circular DNA: A ...<|separator|>
  76. [76]
    Regulated Formation of Extrachromosomal Circular DNA Molecules ...
    This specific DNA population is not detected in the male or female germ cells and is dramatically reduced in later developmental stages and in adult tissues.
  77. [77]
    V(D)J Recombination: Recent Insights in Formation of the ...
    Apr 29, 2022 · V(D)J recombination is an essential mechanism of the adaptive immune system, producing a diverse set of antigen receptors in developing lymphocytes.Missing: ecDNA | Show results with:ecDNA
  78. [78]
  79. [79]
    Coevolving Plasmids Drive Gene Flow and Genome Plasticity ... - NIH
    Nov 5, 2020 · Plasmids are important in microbial evolution and adaptation to new environments. Yet, carrying a plasmid can be costly, and long-term ...<|separator|>
  80. [80]
    Mechanisms Orchestrating Mitochondrial Dynamics for Energy ...
    In this review, we will highlight the molecular pathways involved in the regulation of mitochondrial dynamics and their roles in maintaining energy homeostasis.
  81. [81]
    EccDNA atlas in male mice reveals features protecting genes ... - NIH
    Feb 22, 2025 · Here, we present an atlas of eccDNA across seven tissues of male mice spanning four ages. EccDNA correlates with open chromatin characterized by signatures of ...
  82. [82]
    Chloroplasts and Photosynthesis - Molecular Biology of the Cell
    Chloroplasts perform photosynthesis during the daylight hours. The immediate products of photosynthesis, NADPH and ATP, are used by the photosynthetic cells to ...
  83. [83]
    Persistent Viral Infections - Medical Microbiology - NCBI Bookshelf
    Persistent infections are characterized as those in which the virus is not cleared but remains in specific cells of infected individuals.
  84. [84]
    The unresolved role of mitochondrial DNA in Parkinson's disease
    Among these deletions is the 'common deletion': a 4 977-base-pair deletion located between the MT-ATP8 and MT-ND5 genes as indicated by the dark green arrows.
  85. [85]
    mtDNA Maintenance and Alterations in the Pathogenesis of ...
    Among PD patients, the most common mtDNA deletion is the mtDNA 4977-bp deletion, involving the gene encoding the complex I subunit [129].
  86. [86]
    Epstein-Barr Virus (EBV): Biology and Clinical Disease - PMC
    During the latent phase, the virus is present in the nucleus as a circular episome tethered to the host genome chromatin utilizing a viral protein named EBNA-1.
  87. [87]
    HIV-1 latency and virus production from unintegrated genomes ...
    HIV-1 integration is prone to a high rate of failure, resulting in the accumulation of unintegrated viral genomes (uDNA) in vivo and in vitro.
  88. [88]
    PTEN Mediates the Silencing of Unintegrated HIV-1 DNA - PMC - NIH
    Feb 14, 2024 · The persistence and silencing of unintegrated HIV-1 DNA suggest these viral DNA molecules may contribute to HIV-1 latency—the major obstacle to ...
  89. [89]
    Integration of mtDNA pseudogenes into the nuclear genome ... - NIH
    The insertion of a NUMT is a rare event. Hundreds of pseudogenes have been cataloged in the human genome. NUMTs are, in essence, a special type of mutation with ...
  90. [90]
    Mitochondrial Nuclear Retrograde Regulator 1 (MNRR1) rescues ...
    Nov 30, 2020 · MELAS is a multisystem encephalomyopathy disorder that can result from a heteroplasmic mutation in the mitochondrial DNA (mtDNA; m.3243A > G) at ...<|separator|>
  91. [91]
    Genome-wide characterization of extrachromosomal circular DNA in ...
    We characterized the hallmarks of plasma eccDNA in SLE patients, including the lower normalized number and GC content of eccDNA in SLE plasma than in the ...Missing: studies inflammation conditions
  92. [92]
    Plasmid carriage can limit bacteria–phage coevolution - PMC - NIH
    Our findings suggest that combined exposure to both phages [32] and plasmids [33] in Pseudomonas chronic infections could exacerbate selection for mucoidy, ...
  93. [93]
    Comprehensive analysis of eccDNA characteristics and associated ...
    Mar 20, 2025 · The cyclic structure of eccDNA and its function in autoimmune diseases make it a potential biomarker for SLE. A total of 23 ISLE patients, 20 ...
  94. [94]
  95. [95]
  96. [96]
    Extrachromosomal DNA–Driven Oncogene Dosage Heterogeneity ...
    Extrachromosomal DNA (ecDNA) amplification enhances intercellular oncogene dosage variability and accelerates tumor evolution by violating foundational ...
  97. [97]
    Extrachromosomal circular DNA in cancer drug resistance and its ...
    Amplification of oncogenes and drug resistance genes in ecDNA promotes tumorigenesis and drug resistance. Many studies in recent years have confirmed that ...
  98. [98]
    MYC ecDNA promotes intratumour heterogeneity and plasticity in ...
    Mar 12, 2025 · Live-cell imaging shows uneven segregation of extrachromosomal DNA elements and transcriptionally active extrachromosomal DNA hubs in cancer.
  99. [99]
  100. [100]
    Coordinated inheritance of extrachromosomal DNAs in cancer cells
    Nov 6, 2024 · ecDNAs exhibit aggressive behaviour in cancer cells as they can rapidly shift in copy number and evolve novel gene regulatory relationships.