Fact-checked by Grok 2 weeks ago

Homologous recombination

Homologous recombination () is a highly conserved genetic process that enables the precise exchange of DNA sequences between two similar or identical DNA molecules, serving as a key mechanism for repairing double-strand breaks (DSBs) and other complex DNA lesions while promoting . This high-fidelity pathway uses an undamaged homologous DNA template—typically a sister chromatid in cells or a during —to restore the original genetic information, thereby maintaining genomic integrity and preventing deleterious mutations. The mechanism of HR is tightly regulated and begins with the detection and processing of DNA damage, such as a DSB, which triggers end resection by nucleases to generate 3'-single-stranded DNA (ssDNA) overhangs. These ssDNA tails are coated with recombinase proteins like RAD51 (in eukaryotes), forming nucleoprotein filaments that perform a homology search and invade the homologous duplex DNA, creating a displacement loop (D-loop). Subsequent steps involve to extend the invading strand, branch migration, and resolution of Holliday junctions either via crossover (exchanging large segments) or non-crossover outcomes, such as synthesis-dependent strand annealing, ensuring error-free repair. This process is facilitated by accessory proteins like RAD54 for filament stabilization and helicase for junction dissolution, with regulation varying between mitotic and meiotic contexts to control crossover frequency. Beyond repair, HR plays pivotal roles in cellular and organismal biology. In somatic cells, it counters DSBs caused by endogenous replication fork collapse, ionizing radiation, or chemotherapeutic drugs, suppressing tumorigenesis by avoiding error-prone alternatives like non-homologous end joining. During meiosis, HR is essential for chiasma formation, which physically links homologous chromosomes to ensure their proper segregation in the first meiotic division, producing balanced haploid gametes. Meiotic HR also shuffles alleles through crossovers, generating genetic variation that drives evolution and adaptation in sexually reproducing organisms. Dysregulation or deficiency in HR pathways contributes to genomic instability, linking defects in genes like BRCA1/2 or FANCA to hereditary cancers and syndromes such as Fanconi anemia.

History

Early Observations

In 1913, Alfred Sturtevant conducted pioneering genetic mapping studies using recombination frequencies observed in Drosophila melanogaster, demonstrating that genes are arranged linearly on chromosomes and that the frequency of recombination between them correlates with their physical distance. By analyzing crosses involving sex-linked traits such as white eyes, miniature wings, and yellow body color, Sturtevant constructed the first genetic linkage map, establishing the concept of genetic linkage and the role of crossing over in generating recombinant progeny. This work provided early evidence for homologous recombination as a mechanism enabling genetic exchange during meiosis. In 1928, Frederick Griffith's experiments with revealed the phenomenon of bacterial transformation, where heat-killed virulent (smooth) pneumococci could transfer genetic material to live non-virulent (rough) strains, enabling the latter to acquire and cause in mice. Griffith observed that mixing heat-killed type II smooth bacteria with live type III rough bacteria resulted in the recovery of live type II smooth bacteria from infected mice, indicating a stable heritable change induced by a transferable factor. This discovery highlighted the possibility of genetic material transfer between homologous bacterial strains, laying foundational observations for homologous recombination in prokaryotes. Building on Griffith's findings, , Colin MacLeod, and confirmed in 1944 that deoxyribonucleic acid (DNA) was the transforming principle responsible for these genetic transfers in pneumococci. Through purification and biochemical assays, they demonstrated that DNA extracted from virulent type III pneumococci could irreversibly transform non-virulent type II strains into stable virulent type III variants, resistant to enzymatic degradation by proteases and ribonucleases but inactivated by deoxyribonucleases. Their experiments established DNA as the heritable substance capable of mediating homologous genetic exchange, shifting understanding toward nucleic acids as carriers of genetic information. In 1931, Harriet Creighton and provided cytological evidence for meiotic crossing over in Zea mays (), correlating with physical chromosome exchanges. Using a strain with a chromosomal knob and a translocation on , they showed that progeny exhibiting recombinant genotypes for the C (colored) and Wx (waxy) loci also displayed corresponding cytological rearrangements, such as knob translocations, confirming that genetic crossing over involves actual breakage and rejoining of homologous chromosomes. This study offered direct proof that homologous recombination occurs at the physical level during in eukaryotes.

Molecular Elucidation

In the mid-20th century, pivotal genetic experiments illuminated the molecular underpinnings of homologous recombination through studies of bacterial conjugation. François Jacob and Élie Wollman demonstrated in the 1950s that genetic recombination occurs during the transfer of chromosomal material from donor to recipient bacteria in Escherichia coli, using interrupted mating techniques to map the sequential entry of genes and reveal the role of physical DNA transfer in generating recombinant progeny. These findings established recombination as a process involving the exchange of homologous DNA segments, building on earlier observations of genetic transformation in bacteria. A major breakthrough came in 1965 with the identification of the protein in E. coli by Alvin J. Clark and Anne D. Margulies, who isolated recombination-deficient mutants and showed that the recA gene is essential for , thereby linking specific genetic loci to the machinery of DNA strand exchange. Concurrently, in 1964, Robin Holliday proposed a seminal model for recombination in fungi, positing the formation of a four-way DNA junction—now known as the —as a key intermediate that facilitates strand breakage, invasion, and resolution to produce recombinant molecules and explain phenomena like gene conversion. Building on these ideas, and Charles M. Radding refined the model in 1975 by introducing an asymmetric mechanism of strand invasion, where a single-stranded DNA segment from one duplex invades a homologous duplex, displacing a strand and initiating heteroduplex formation without requiring symmetrical nicks on both molecules. This framework provided a more precise biochemical explanation for the of recombination and influenced subsequent understandings of repair processes.

Basic Principles

Definition and Overview

Homologous recombination (HR) is a conserved genetic process that facilitates the exchange of DNA sequences between two similar or identical molecules, relying on regions of homology to align and swap genetic material. This mechanism enables the precise transfer of information from an undamaged template to repair or modify damaged DNA, distinguishing it as a high-fidelity pathway essential for genomic stability. HR occurs universally across all domains of life, including bacteria, archaea, and eukaryotes, as well as in viruses, underscoring its ancient evolutionary origins and fundamental role in cellular function. HR serves several critical biological functions, primarily the repair of double-strand breaks (DSBs) in DNA, the promotion of genetic diversity during meiosis, and the integration of horizontally transferred genes. In DSB repair, HR accesses an intact homologous sequence to accurately restore the original genetic information, thereby minimizing mutations and preserving genome integrity during replication or environmental stress.30478-2) During meiotic recombination in eukaryotes, HR drives reciprocal exchanges between homologous chromosomes, ensuring proper segregation and generating novel allele combinations that enhance genetic variation in offspring. In bacteria, HR facilitates horizontal gene transfer by allowing the stable incorporation of exogenous DNA with sufficient sequence similarity into the host genome, thereby expanding adaptive capabilities. Unlike (NHEJ), an intrinsically error-prone pathway that ligates DSB ends with minimal homology—often introducing insertions or deletions—HR depends on a homologous template for accurate sequence reconstruction, making it the preferred mechanism for precise repair. The core process of HR initiates with a DSB, followed by 5' end resection to produce 3'-overhanging single-stranded DNA tails coated by recombinase proteins. These tails invade a homologous duplex, forming a displacement loop and enabling strand exchange; subsequent branch migration extends the heteroduplex region, and resolution of intermediates (such as Holliday junctions) completes the exchange or repair.

Core Molecular Mechanism

Homologous recombination (HR) primarily serves to repair double-strand breaks (DSBs) in DNA by utilizing a homologous sequence as a template for accurate repair. The process begins with the recognition and processing of the DSB, where the 5' ends of the broken DNA strands undergo resection by a combination of helicases and nucleases, generating extended 3' single-stranded DNA (ssDNA) overhangs. This resection step, extending hundreds to thousands of nucleotides, is essential for exposing sequences that can search for homology and committing the break to the HR pathway. The ssDNA tails are then bound by recombinase proteins from the RecA family, which polymerize to form a helical nucleoprotein filament that stabilizes the ssDNA and enables an active search for homologous duplex DNA. This presynaptic filament facilitates strand invasion, in which one of the 3' ssDNA ends invades the intact homologous duplex, pairing with the complementary strand and displacing the non-complementary strand to form a displacement loop (D-loop). The D-loop is a critical early intermediate, characterized by a bubble-like structure where the invading ssDNA anneals to one strand of the duplex while displacing the other, creating a primer-template junction for repair synthesis. Using the homologous duplex as a template, DNA polymerase extends the invading 3' end through new DNA synthesis, copying the genetic information to restore the broken sequence. The process proceeds to form a , a key branched intermediate consisting of a cross-shaped DNA structure where two homologous duplexes are joined by reciprocal strand exchanges, allowing for the potential transfer of alleles between chromosomes. First conceptualized by Robin Holliday in 1964, the features two continuous (non-crossed) arms and two crossed arms formed by single-stranded connections, enabling branch migration—a sliding of the junction along the DNA to extend the heteroduplex region. The second DSB end may then be captured, leading to a double , which undergoes resolution by structure-specific endonucleases that cleave the junctions in orientations yielding either crossover (exchange of flanking sequences) or non-crossover products (gene conversion without exchange). The , proposed by Szostak et al. in 1983, formalized these steps as a canonical pathway for HR-mediated repair. In genetic mapping, the frequency of HR events between linked loci quantifies their physical proximity, with recombination frequency (RF) calculated as RF = (number of recombinant progeny / total progeny) × 100%, expressed in centimorgans (cM); values below 50% indicate linkage, as crossovers occur probabilistically along the chromosome.

In Bacteria

RecBCD Pathway

The RecBCD pathway is the primary mechanism of homologous recombination in wild-type Escherichia coli for repairing double-strand breaks (DSBs) in DNA. The RecBCD enzyme complex, composed of three subunits (RecB, RecC, and RecD) forming a 330-kDa heterotrimer, binds to blunt or nearly blunt DSB ends and exhibits both ATP-dependent helicase activity to unwind DNA and nuclease activity to degrade strands. This complex processes the DNA end by unwinding the duplex at a rate of approximately 300–1,000 base pairs per second while simultaneously resecting the 5' strands, generating a 3' single-stranded DNA (ssDNA) tail essential for downstream recombination. Degradation continues processively until the enzyme encounters a Chi site, an octameric sequence (5'-GCTGGTGG-3') present approximately once every 5 kb in the E. coli genome, with over 1,000 sites oriented predominantly toward the replication origin. Upon recognition by the RecC subunit, which scans the ssDNA, the complex undergoes a conformational change that attenuates its activity on the 3' strand, nicking the 5' strand about 5 nucleotides 3' to the site and thereby protecting the emerging 3' ssDNA tail from further degradation. This switch promotes the loading of protein onto the 3' ssDNA tail by the complex, forming a filament that facilitates strand invasion into a homologous duplex DNA template. The pathway is crucial for repairing DSBs induced by , mechanical stress during replication fork collapse, or other genotoxic events, ensuring genomic integrity and cell viability. Additionally, integrates with natural genetic exchange processes, processing linear donor DNA fragments during conjugation and to enable their recombination into the recipient ; in wild-type E. coli, it mediates approximately 99% of such recombination events. Experimental evidence from genetic studies underscores the pathway's essential role. Mutants lacking functional RecB or RecC subunits (e.g., recB or recC null strains) exhibit severely reduced recombination proficiency, hypersensitivity to DNA-damaging agents like UV or , and compromised viability, with survival rates dropping to around 30% under normal conditions due to unrepaired DSBs. In contrast, recD mutants retain recombination activity but display hyper-recombination and reliance on alternative nucleases like RecJ, highlighting RecD's specific role in enhancing 5' strand degradation prior to . These findings, derived from conjugation and assays, confirm that is indispensable for DSB-initiated recombination in E. coli.

RecF Pathway

The RecF pathway represents an alternative route for homologous recombination in , primarily dedicated to the repair of single-stranded DNA (ssDNA) gaps rather than double-strand breaks (DSBs). Central to this pathway is the RecFOR complex, composed of the RecF, RecO, and RecR proteins, which cooperatively bind at ssDNA-dsDNA junctions to process gaps and facilitate the assembly of filaments. The tetrameric RecR forms a ring-like structure that encircles double-stranded DNA (dsDNA) and interacts with RecF, while RecO stabilizes the complex by engaging ssDNA coated with single-stranded DNA-binding protein (), thereby displacing to enable loading. This mediator function is essential for initiating strand invasion during recombination, particularly in scenarios involving replication-associated damage. The pathway is prominently activated in RecBC-deficient mutants of , where the canonical pathway is impaired, allowing RecF-dependent mechanisms to compensate for recombination proficiency. It also plays a key role in repairing UV-induced lesions, such as , by stabilizing stalled replication forks and promoting gap repair downstream of . In wild-type cells, the RecF pathway integrates with post-replication repair processes, where it addresses daughter-strand gaps arising from blocked replication forks, and contributes to the response by forming filaments that activate LexA repressor cleavage, inducing DNA damage tolerance genes including those for translesion synthesis. Unlike the pathway, which features potent and activities for DSB resection, the RecF route lacks intrinsic function and instead emphasizes gap filling through -mediated strand exchange, often relying on auxiliary helicases like RecQ for ssDNA extension. Genetic studies in bacterial mutants provide strong evidence for the RecF pathway's contributions, demonstrating elevated recombination frequencies in RecBC-deficient backgrounds when RecF, RecO, or RecR functions are intact, such as in recB sbcB strains where conjugational recombination is restored to near-wild-type levels. Conversely, disruptions in recF, recO, or recR in these compensatory mutants lead to severe recombination defects, underscoring the pathway's specificity for gap repair and its non-redundancy with under standard DSB conditions. These findings highlight the RecF pathway's adaptive role in maintaining genome stability during replication stress.

In Eukaryotes

Mitotic Recombination

Homologous recombination () during the mitotic in eukaryotes primarily serves to repair DNA double-strand breaks (DSBs) and maintain stability in somatic cells, utilizing the sister chromatid as the preferred template to restore genetic information accurately. This process is essential for repairing damage arising from endogenous sources, such as replication , or exogenous agents, ensuring faithful segregation and preventing that could lead to diseases like cancer. Unlike meiotic HR, mitotic HR favors non-crossover outcomes to minimize structural alterations in the . Mitotic HR is predominantly active during the S and G2 phases of the , when has generated identical available as homologous templates for repair. This temporal restriction ensures that HR can access undamaged donor sequences, as G1-phase cells lack sisters and rely more on . The process begins with the detection of DSBs, followed by end resection to generate single-stranded DNA (ssDNA) overhangs, which are coated by recombinase proteins to facilitate homology search and strand exchange. Chromatin modifications play a critical role in signaling DSBs for HR repair in mitotic cells. Phosphorylation of histone H2AX to form γH2AX marks the sites of DSBs, spreading over megabases to create foci that recruit repair factors and facilitate for accessibility. This modification, mediated by kinases like , coordinates the DNA damage response and promotes the transition to HR by aiding in the recruitment of resection machinery. Key proteins in mitotic HR include the MRN complex (MRE11-RAD50-NBS1), which initiates DSB end resection by unwinding and degrading DNA ends to produce the 3' ssDNA tails necessary for downstream steps. Following resection, the recombinase RAD51, the eukaryotic homolog of bacterial , forms nucleoprotein filaments on ssDNA to drive strand invasion into the homologous sister chromatid, initiating repair synthesis. In human cells, RAD51 not only mediates this invasion but also protects under-replicated DNA during , supporting completion of replication and reducing breakage. To preserve genome stability, mitotic HR suppresses crossover products, which could lead to loss of heterozygosity (LOH) and unmask recessive mutations or cause chromosomal rearrangements. This bias toward non-crossover resolutions, such as gene conversion or break-induced replication without exchange, is enforced by anti-crossover factors like SRS2 helicase and the BLM-TOP3A-RMI1/2 (BTR) complex, which dissolve joint molecules to yield non-crossover outcomes. Defects in this suppression can promote LOH, contributing to tumorigenesis. Examples of mitotic HR include the repair of collapsed replication forks, where stalled forks generate one-ended DSBs that are resected and invaded by RAD51 filaments using the sister chromatid to restart replication and fill gaps. Similarly, DSBs induced by chemotherapeutics, such as or inhibitors, are preferentially repaired by HR in S/G2 phases to avoid error-prone alternatives, with RAD51 recruitment being crucial for cell survival post-treatment.

Meiotic Recombination

Meiotic recombination is a specialized form of homologous recombination that occurs during I of in eukaryotes, playing a crucial role in generating through the exchange of genetic material between and ensuring accurate segregation. This process begins with the formation of programmed DNA double-strand breaks (DSBs) induced by the topoisomerase-like Spo11, which creates breaks at specific sites. These DSBs undergo end resection to generate 3' ssDNA overhangs, coated by the meiosis-specific DMC1 along with RAD51 to form nucleoprotein filaments that perform homology search and initiate strand invasion into the homologous chromosome for repair. Unlike , which primarily repairs accidental damage, meiotic recombination is tightly regulated to produce a limited number of crossovers per chromosome pair, typically one to a few, to promote diversity while avoiding excessive fragmentation. DSB formation hotspots are preferentially located in open regions, such as promoters, and their distribution is influenced by architecture and epigenetic marks, with Spo11 activity requiring accessory proteins like Rec102/Rec114/Mre11 (in ) to target these sites. The (), a proteinaceous that forms between homologous during zygotene and pachytene stages, stabilizes and facilitates the progression of recombination intermediates, while also imposing spatial constraints that help regulate crossover placement. Within the , recombination nodules mark sites of active recombination, and the complex's liquid-crystalline properties contribute to the even spacing of crossovers by influencing DSB processing and repair pathway choices. The promotion of crossovers is achieved through a designation process that channels certain recombination intermediates into crossover outcomes, while others are directed to noncrossover pathways, ensuring an obligate crossover per pair in many organisms. Crossover , a where one crossover reduces the likelihood of another nearby, operates over long distances (often genome-wide) and is mediated by the SC and associated proteins, resulting in evenly spaced chiasmata. Key meiosis-specific proteins include the MutS homologs MSH4 and MSH5, which form a heterocomplex that stabilizes joint molecules (such as double Holliday junctions) early in recombination and promotes the bias toward crossovers by interacting with other ZMM (Zip1/2/3, Msh4/5, Mer3) proteins. Later, the MutL homologs MLH1 and MLH3 form an endonuclease complex that resolves these joints into crossovers, with MLH1-MLH3 foci appearing at designated sites and requiring prior MSH4-MSH5 loading for recruitment. The ultimate outcome of meiotic recombination is the formation of chiasmata, physical manifestations of crossovers that hold homologous chromosomes together until I, thereby ensuring proper bipolar and preventing . In mutants lacking MLH1 or MLH3, crossover numbers are severely reduced, leading to univalents and meiotic failure, underscoring their essential role in chiasma formation. This regulated process thus balances genetic diversity with genomic stability across eukaryotic species.

Recombination Models

Double-Strand Break Repair Model

The double-strand break repair (DSBR) model, proposed in 1983, describes a mechanism of homologous recombination in which a double-strand break (DSB) in DNA is repaired using a homologous template, involving both ends of the break to form a double Holliday junction intermediate. This model posits that recombination initiates with the formation of a DSB, followed by extensive 5' to 3' resection of the broken ends to generate long 3'-single-stranded DNA (ssDNA) tails. These tails are coated by the recombinase RAD51 (or RecA in bacteria), forming a nucleoprotein filament that facilitates search for and invasion of a homologous duplex DNA sequence, creating a displacement loop (D-loop). Following initial strand invasion, extends the invading 3' end using the homologous template, while the second free end of the DSB is captured and anneals to the displaced strand in the , leading to the formation of a second . Ligation of nicks results in a double (dHJ) structure, which represents a key symmetric intermediate linking the broken DNA molecule to the intact homolog. The dHJ can then be resolved by structure-specific endonucleases, such as GEN1 (or its yeast homolog Yen1), which introduce coordinated incisions on opposite strands, yielding either crossover or non-crossover products with equal probability if resolution is random. Alternatively, dHJs may be dissolved by helicase-topoisomerase complexes like BLM-TOP3A-RMI1/2 to produce exclusively non-crossover outcomes. The DSBR model is particularly prominent in meiotic recombination, where it promotes crossover formation essential for proper chromosome segregation, but it occurs less frequently in mitotic cells, where non-crossover repair predominates to minimize genomic rearrangements. Unlike models involving single-end , DSBR distinctly engages both DSB ends, enabling the potential for reciprocal exchange of flanking markers. Experimental validation in has included electron (EM) visualization of recombination intermediates, such as joint molecules with paired equal-length arms consistent with Holliday junctions, and yeast two-hybrid assays confirming protein interactions (e.g., RAD51 with mediators like RAD52) critical for filament formation and strand exchange. These studies, including direct observation of dHJ-like structures , have substantiated the model's predictions for eukaryotic recombination.

Synthesis-Dependent Strand Annealing

Synthesis-dependent strand annealing (SDSA) is a non-crossover mechanism of homologous recombination that repairs double-strand breaks (DSBs) through unidirectional strand invasion and subsequent annealing, primarily in eukaryotic cells. In this pathway, following DSB formation and 5' to 3' resection of the broken ends to generate 3' single-stranded DNA (ssDNA) tails, one resected end invades a homologous donor sequence, forming a facilitated by the filament. DNA then extends the invading 3' end using the donor as a template, copying genetic information to restore the damaged sequence. The extended invading strand is subsequently displaced from the donor and anneals to the other resected end of the original break, followed by gap filling and ligation to complete repair. Unlike pathways involving double Holliday junctions, SDSA does not form stable joint molecules that could lead to crossover products, making it the predominant mode of DSB repair in mitotic cells to preserve stability without allelic exchanges. This crossover avoidance is promoted by helicases such as Srs2 in , which disrupt extended D-loops to favor annealing over second-end capture. SDSA plays a key role in repairing two-ended DSBs arising from collapsed replication forks, where both ends are available for annealing after synthesis, and contributes to telomere maintenance by facilitating noncrossover repair of telomeric DSBs without disrupting ends. Evidence for SDSA emerged from studies in , where P-element-induced gap repair demonstrated efficient copying of ectopic donor sequences into DSB sites, resulting in gene conversion tracts without associated crossovers, consistent with unidirectional synthesis and annealing. In , assays using split donor templates on separate chromosomes revealed noncrossover gene conversion products, with repair kinetics and tract lengths supporting SDSA as a major mitotic pathway that channels most DSBs away from crossover-prone intermediates. These findings highlight SDSA's preference for short-tract gene conversion in standard repairs but allow for long-tract conversion when extensive occurs, such as during replication fork restart.

Single-Strand Annealing

Single-strand annealing () is a homology-directed DNA double-strand break (DSB) repair pathway that operates independently of RecA homologs like RAD51, relying instead on homologous direct repeats flanking the break site to rejoin DNA ends. This process is prominent in eukaryotes and serves as an error-prone alternative to canonical homologous recombination, particularly in regions with repetitive sequences. The mechanism of SSA begins with the resection of DSB ends by nucleases such as CtIP and EXO1, generating long 3' single-stranded DNA (ssDNA) tails that expose complementary sequences within direct repeats. These ssDNA tails then anneal via base-pairing, facilitated by , which forms ring structures that bind and align the homologous regions without requiring strand invasion. Following annealing, non-homologous flaps—resulting from asymmetric resection—are cleaved by the ERCC1-XPF endonuclease, and the processed ends are ligated by I or III, completing the repair. This pathway was first described in models, where it was shown to mediate repair between homologous repeats. The outcome of SSA is invariably non-conservative, leading to the deletion of the intervening sequence between the annealing repeats and any non-homologous tails, with no transfer of genetic information from a donor template. Unlike gene conversion in homologous recombination, SSA does not restore the original sequence and thus promotes loss-of-heterozygosity or genomic rearrangements. SSA is particularly active in genomic contexts enriched with direct repeats, such as Alu elements (short interspersed nuclear elements) or transposon-flanked regions, where it can facilitate rapid repair but at the cost of structural alterations. It functions as an error-prone backup to homologous recombination, especially during the S/G2 phases of the when resection is extensive. Evidence for SSA's role and regulation comes from studies in mammalian cells, where its frequency increases substantially upon disruption of RAD51, shifting repair away from canonical homologous recombination toward annealing-based pathways. For instance, RAD51 knockdown in ovary cells elevated SSA events by approximately 11%, highlighting its compensatory activation. In vitro assays and structural analyses further confirm RAD52's annealing efficiency on ssDNA substrates, underscoring its central, invasion-independent function in eukaryotes.

Break-Induced Replication

Break-induced replication (BIR) is a specialized pathway of homologous recombination () in eukaryotes dedicated to repairing one-ended double-strand breaks (DSBs), such as those arising from collapsed replication forks or unprotected ends. Unlike bidirectional repair mechanisms, BIR initiates unidirectional from the invading broken end into a homologous template, often leading to the duplication of large chromosomal segments. This process is conserved across eukaryotes and plays a critical role in maintaining under conditions where traditional two-ended DSB repair is unavailable. The mechanism of BIR commences with the 5' to 3' resection of the broken DNA end, generating a long 3' single-stranded DNA (ssDNA) overhang that is initially bound by replication protein A (RPA). This overhang is then remodeled into a nucleoprotein filament by the RAD51 recombinase, which facilitates the search for and single-end invasion of a homologous sequence on a sister chromatid or homologous chromosome. The invasion establishes a displacement loop (D-loop) intermediate, where the 3' end of the invading strand serves as a primer for DNA polymerase-mediated extension. Following D-loop formation, semi-conservative DNA replication is initiated, primarily driven by DNA polymerase delta (POLD) for both leading- and lagging-strand synthesis, with DNA polymerase epsilon (POLE) contributing to the leading strand and regulatory functions. This replication fork migrates processively toward the chromosome end, copying the template for potentially hundreds of kilobases at a rate slower than normal S-phase replication (approximately 0.5 kb/min in yeast). BIR outcomes typically include long-tract non-crossover gene conversion, where the repaired arm is extensively replaced by the homologous , and conservative of the newly synthesized to one daughter cell. However, the process can also promote copy number changes through template switching or half-crossovers, leading to duplications, deletions, or translocations. In specific cellular contexts, BIR facilitates the restart of stalled or collapsed replication forks during , elongation of telomeres in telomerase-deficient cells, and resolution of dicentric bridges by copying until a natural end is reached. For instance, in mutants lacking , BIR repairs critically shortened telomeres, resulting in survivor cells with elongated tracts. Evidence for BIR has been robustly established through yeast-based assays monitoring , where inducible DSBs near telomeres BIR-dependent lengthening observable via Southern blotting and . In cancer studies, BIR signatures manifest as amplifications and copy number variations in tumor cells, particularly in alternative lengthening of telomeres ()-positive cancers, linking the pathway to oncogenesis and genomic . These findings underscore BIR's dual role in survival and mutagenesis.

In Viruses

General Mechanisms

Homologous recombination (HR) plays a pivotal role in viral replication by enabling the repair of DNA lesions and the restart of stalled replication forks, which is essential for efficient genome duplication in DNA viruses. This mechanism allows viruses to maintain genome integrity amid host-imposed stresses, such as nucleotide shortages or antiviral nucleases. In RNA viruses, analogous recombination events facilitate template switching during replication, supporting continuous viral propagation. Viruses harness HR to generate genetic diversity, promoting rapid evolution in both DNA and RNA viruses by creating chimeric genomes from co-infecting parental strains. This diversity aids in evading host defenses, including innate immune responses and RNA interference, by producing variants that resist restriction factors or antibodies. Many viruses rely on host HR factors for these processes; for instance, RAD52 facilitates strand annealing and exchange in herpesvirus replication. Certain viral polymerases also incorporate HR-like functions, such as copy-choice recombination, where the polymerase dissociates and reassociates with a homologous template to complete synthesis. In contrast to the precise, error-minimizing cellular , viral HR is often asymmetric—favoring single-strand exchanges—and integrated with high rates to accelerate . This is particularly evident in single-stranded viruses, such as parvoviruses, where second-strand synthesis is initiated by the viral genome's terminal hairpin serving as a primer for host DNA polymerases. Core strand invasion, a conserved step in HR, is adapted in viruses to initiate these exchanges efficiently. Experimental quantification of viral HR typically employs assays involving co-infection of marker-bearing viral strains, followed by detection of recombinant progeny via sequencing or phenotypic selection.

Examples in Specific Viruses

In (HSV), a , homologous recombination facilitates viral genome replication and repair by utilizing viral proteins that mimic host recombination machinery. The viral UL12 performs end resection to generate single-stranded DNA overhangs, enabling strand invasion, while the single-strand ICP8 stabilizes these intermediates and promotes annealing during recombination. This process allows HSV to co-opt the host pathway, including RAD51, for efficient recombination-dependent replication. Among RNA viruses, (IAV) employs an HR-like mechanism through template switching during replication, which contributes to genetic diversity beyond classical reassortment of its segmented genome. The viral (RdRp) pauses and switches templates based on , generating intra-segmental recombinants that can alter virulence or host adaptation, as observed in historical strains like the 2009 H1N1 virus. This template-switching recombination occurs at rates influenced by fidelity and is distinct from but complements it in driving . In human immunodeficiency virus (HIV), homologous recombination manifests during reverse transcription via copy-choice strand transfers mediated by the viral reverse transcriptase. This enzyme switches templates between homologous regions of the two RNA genomes in the virion, producing recombinant progeny that contribute to immune escape and drug resistance; for instance, strand transfers occur at frequencies up to 10% per replication cycle in dual infections. Such events are essential for generating the full-length proviral DNA, integrating homologous sequences while occasionally introducing deletions or insertions. Coronavirus recombination, exemplified by , relies on RdRp-mediated template switching to produce chimeric genomes, particularly during co-infections that fueled variant emergence post-2020. In cases of and co-circulation, recombinant strains like XBB lineages arose from template switches in the spike gene, enhancing transmissibility; these events occur via the polymerase pausing at homologous sites and jumping to alternative templates, with recombination hotspots identified in non-structural protein regions. This mechanism has driven the generation of variants of concern, such as sublineages, by combining from divergent lineages. Picornaviruses, positive-sense single-stranded viruses like , utilize homologous recombination to generate defective interfering () particles that attenuate and modulate host responses. During high-multiplicity infections, the RdRp engages in template switching at homologous sequences, producing deleted genomes that package into virions and interfere with full-length synthesis by competing for . These particles, formed at rates exceeding 1% in , represent a natural antiviral strategy, as recombination rates correlate with polymerase error proneness. Recent studies as of 2025 highlight homologous recombination in es, such as , where low-fidelity polymerases promote template switching to overproduce DI particles, potentially contributing to vaccine escape in live-attenuated formulations. In replicon-based , recombination events generate defective genomes that reduce over passages, as seen in serial propagation experiments; this underscores the role of HR in limiting vaccine stability and informs design of next-generation self-amplifying platforms.

Consequences of Dysfunction

Genetic Instability

Defects in homologous recombination () can lead to the persistence of unresolved recombination intermediates, such as D-loops or Holliday junctions, which fail to be properly dissolved or resolved during . These intermediates create covalent links between chromatids that persist into , resulting in ultra-fine bridges that are prone to breakage and subsequent fragmentation. Ectopic HR, where recombination occurs between non-allelic homologous sequences, often promotes (LOH) by converting heterozygous regions to homozygous states through gene conversion or break-induced replication, thereby reducing genetic diversity and increasing susceptibility to further mutations. Such HR failures contribute to various chromosomal aberrations, including translocations that arise when erroneous repair pathways like single-strand annealing (SSA) or break-induced replication (BIR) process double-strand breaks in repetitive genomic regions. typically results in large deletions by annealing complementary sequences after flap removal, while BIR can cause extensive copy number variations and inversions, particularly in telomere-proximal repeats. In repeat-rich areas, these mechanisms exacerbate instability by favoring non-conservative outcomes over precise . Mutations disrupting HR components, such as in the yeast RAD52 gene, primarily impair HR, leading to reduced recombination and synthetic lethality when combined with defects in other repair factors like BRCA2. This synthetic lethality underscores the interconnectedness of HR with genome maintenance, where dual impairments overwhelm cellular repair capacity and amplify instability. HR defects manifest differently in somatic versus germinal cells: somatic instability often accumulates through replication stress and environmental damage, leading to progressive aberrations, while germinal defects disrupt meiotic processes and may contribute to transgenerational instability. Both contexts link to aging, as declining HR efficiency with age impairs double-strand break repair, fostering cumulative genomic damage. In HR-deficient cells, this is quantitated by elevated micronuclei formation, indicative of breakage and missegregation, and altered sister chromatid exchanges (), where reduced HR shifts reliance to error-prone alternatives. For instance, HR impairment can increase micronuclei by up to several-fold in response to replication stress, highlighting the pathway's role in preventing .

Associated Diseases

Dysfunction in homologous recombination () pathways is implicated in several human diseases, primarily through increased genomic that leads to , developmental defects, or oncogenic transformations. Genetic instability phenotypes, such as chromosomal aberrations and mutations, arise from impaired HR and manifest in syndromes characterized by heightened cancer risk and other pathologies. (FA) is a rare caused by mutations in genes involved in the FA pathway, which intersects with HR, leading to failure, congenital abnormalities, and a profoundly elevated risk of cancers including and solid tumors. For instance, mutations in FANCD2, a key HR-associated protein, disrupt the repair of interstrand crosslinks and double-strand breaks via HR, resulting in hypersensitivity to DNA damaging agents and progressive exhaustion. This pathway's role in promoting HR fidelity is evidenced by studies showing that FA core complex components, like FANCA and FANCG, facilitate of chromosomal breaks. Deficiencies in and genes, central to HR-mediated double-strand break repair, predispose individuals to hereditary and ovarian cancers due to impaired HR, which causes accumulation of genomic scars such as and structural variants. BRCA1/2 mutations lead to HR deficiency (HRD), a hallmark that distinguishes these tumors and correlates with aggressive phenotypes and poor in ovarian and cancers. In BRCA1/2-deficient cells, unrepaired breaks shift reliance to error-prone repair mechanisms, fostering tumorigenesis. Bloom syndrome, resulting from biallelic mutations in the gene encoding a , features hyper-recombination and genomic instability, manifesting as growth retardation, , and a 150-fold increased cancer risk, particularly leukemias and lymphomas. BLM normally suppresses excessive to maintain genome stability, and its loss elevates sister chromatid exchanges and mutation rates, driving oncogenesis. Meiotic defects in , such as those from , disrupt double-strand break formation essential for pairing and recombination, leading to through production of aneuploid gametes and impaired . In humans, SPO11 variants are associated with non-obstructive in males and premature ovarian insufficiency in females, highlighting HR's critical role in reproductive . Therapeutically, HR deficiencies confer with , which trap PARP on DNA and overwhelm replication forks in HR-impaired cells, selectively killing /2-mutant tumors while sparing normal cells. This approach has revolutionized treatment for HRD-positive ovarian, breast, and prostate cancers, with clinical trials demonstrating prolonged in BRCA-deficient patients. As of 2025, advancements in HRD testing, including genomic scarring assays and AI-driven classifiers from whole slide images, have improved patient stratification for such therapies.

Evolutionary Conservation

RecA Recombinase Family

The RecA recombinase family consists of ancient, evolutionarily conserved proteins that form the core machinery for homologous recombination in , , and eukaryotes, enabling the repair of DNA double-strand breaks and the exchange of genetic information between homologous sequences. These proteins share a common ancestral origin, with the bacterial gene serving as the prototype, while eukaryotic homologs include RAD51 and its meiosis-specific paralog DMC1, and archaeal RadA represents the counterpart in that domain of life. Sequence alignments reveal approximately 20-30% identity in core functional domains across these family members, underscoring their deep despite billions of years of divergence. Structurally, RecA family proteins are filament-forming ATPases characterized by a monomeric core domain that oligomerizes into extended helical nucleoprotein filaments upon binding single-stranded DNA (ssDNA). These filaments exhibit a right-handed helical pitch of approximately 95 Å with 6-7 monomers per turn, as revealed by early crystallographic studies of RecA in the late 1980s and early 1990s. The proteins bind ssDNA with high affinity, preferentially coating it in a 3:1 nucleotide-to-monomer ratio, which stretches and unwinds the DNA to facilitate downstream reactions; this binding is stabilized by interactions involving conserved loops in the DNA-binding site. Crystal structures of the RecA monomer bound to ADP highlight a central ATPase domain flanked by N- and C-terminal extensions that regulate filament dynamics and accessory protein interactions. Functionally, RecA family recombinases nucleate on ssDNA to form a presynaptic filament that actively searches for homologous double-stranded DNA (dsDNA) sequences through a process driven by thermal motion and transient base-pairing probes. Once homology is identified, the filament catalyzes strand exchange, invading the dsDNA to form a displacement loop (D-loop), with energy provided by ATP hydrolysis that powers conformational changes and filament disassembly. This mechanism is exemplified by bacterial RecA, which promotes efficient recombination in vitro over thousands of base pairs, while eukaryotic RAD51 performs analogous roles in mitotic repair, often requiring mediators like RAD52 and RAD54 for filament stabilization—though these are not core recombinases. In meiosis, DMC1 acts as a specialized variant, enhancing interhomolog bias alongside RAD51. Archaeal RadA shares this filament-based homology search and exchange capability, forming similar structures as confirmed by crystallographic snapshots.

Meiosis-Specific Proteins

Meiosis-specific proteins play crucial roles in regulating during , ensuring proper pairing, (SC) formation, and crossover designation to promote accurate . These proteins, distinct from the core machinery, facilitate the transition from double-strand break (DSB) formation to stable interhomolog interactions, thereby enforcing crossover interference and assurance. While RAD51 functions in both mitotic and meiotic recombination for strand invasion, meiosis-specific factors like Spo11, Hop1/Red1, and the ZMM complex impose additional layers of control unique to . Spo11, a topoisomerase-like endonuclease conserved across eukaryotes, initiates meiotic recombination by generating DSBs essential for homolog pairing and crossover formation. Structurally related to archaeal topoisomerase VI subunit A, Spo11 forms a covalent phosphotyrosyl bond with the 5' DNA ends after cleaving both strands, marking the start of repair via homologous recombination. This DSB formation is tightly regulated and occurs specifically in I of , with Spo11 activity dependent on accessory proteins like Rec102, Rec107, and Rec114 in . Mutations in Spo11 abolish DSBs, leading to severe defects in chromosome and , including high rates of and anaphase bridges due to unresolved entanglements. Hop1 and Red1 are key chromosome axis proteins that assemble along meiotic chromosomes to form the structural backbone for and recombination. Hop1, a HORMA domain-containing protein, binds DNA and recruits Red1, which in turn stabilizes the linear axis by interacting with complexes, promoting interhomolog bias in recombination. Together, they facilitate assembly by organizing chromatin loops and suppressing intersister recombination, ensuring DSB repair favors the homolog. In , Hop1-Red1 complexes localize to axial elements prior to formation, and their absence disrupts , reduces DSB levels, and causes chromosome individualization defects. Mutants in Hop1 or Red1 exhibit and bridges, reflecting failures in crossover formation and chromosome disentanglement during segregation. The ZMM complex, comprising Zip1, Zip2, Zip3, Msh4, and Msh5 (along with Zip4, Mer3, and Spo16), stabilizes early recombination intermediates and promotes class I (interfering) crossovers while coordinating SC polymerization. Zip1 serves as the transverse filament of the SC, zipping homologs together and reinforcing designated crossover sites, whereas , Zip3, and Zip4 form a scaffold that recruits other components to DSB hotspots. Msh4 and Msh5, forming a MutSγ heterodimer, recognize and stabilize double Holliday junctions (dHJs), channeling repair toward crossovers rather than noncrossovers. This complex ensures crossover homeostasis by interfering with adjacent events and is essential for SC-dependent interhomolog interactions. ZMM mutants display reduced class I crossovers, fragmented SCs, and segregation errors, including elevated nondisjunction rates and anaphase bridges from persistent recombination intermediates. These meiosis-specific proteins—Spo11, Hop1/Red1, and ZMM components—are absent in and , indicating they evolved in the last eukaryotic common ancestor (LECA) to adapt recombination for and stability. Their emergence likely coincided with the innovation of meiotic DSBs and SC formation, enabling regulated crossing over in the eukaryotic lineage.

Technological Applications

Gene Targeting

Gene targeting leverages (HR) to introduce precise modifications into the genome, utilizing () pathways. In , a donor DNA template with sequences homologous to the regions flanking a double-strand break (DSB) serves as a blueprint for accurate repair, enabling insertions, deletions, or substitutions at specific loci. This process typically involves providing an exogenous donor template, such as a single-stranded (ssODN) or double-stranded DNA (dsDNA) , which contains the desired genetic flanked by arms of 200-800 base pairs to facilitate strand invasion and copying during repair. Early methods for stimulating HR in gene targeting employed engineered nucleases to induce targeted DSBs, thereby promoting HDR over error-prone (NHEJ). nucleases (ZFNs), consisting of DNA-binding domains fused to the FokI endonuclease, were among the first to achieve site-specific DSBs, enabling HR-mediated knock-ins in mammalian cells with efficiencies up to several percent in optimized systems. Similarly, transcription activator-like effector nucleases (TALENs), which use customizable TALE protein arrays for DNA recognition, induce DSBs with high specificity and have been applied to generate HR-based modifications in human cell lines and embryos, often outperforming ZFNs in off-target profile. These tools laid the foundation for precise genome engineering by exploiting the cell's natural HR machinery following DSB induction.30063-4) Despite their precision, HR-based exhibits low efficiency in mammalian cells, typically ranging from 1-10% of edited alleles, primarily due to from dominant NHEJ pathways and restrictions, as is most active in S/G2 phases. Strategies to enhance efficiency include synchronizing cells to these proliferative phases using agents like or hydroxyurea, which can increase HDR rates by 50% or more by aligning DSB induction with HR-competent windows. A landmark application is the creation of mice via HR in embryonic stem cells, pioneered by , where targeting vectors replace or disrupt endogenous genes, enabling the study of loss-of-function phenotypes in whole organisms. In human induced pluripotent stem () cells, HR has facilitated correction of disease-causing mutations, such as those in the DMD gene for , generating isogenic lines for disease modeling and potential autologous therapies.00232-X) Recent advances as of 2025 have integrated CRISPR-Cas9 with HR donor templates to boost therapeutic editing, achieving higher precision for clinical applications like correcting mutations in patient-derived cells. By combining Cas9-induced DSBs with optimized donors and HDR enhancers, such as small-molecule inhibitors of NHEJ, editing efficiencies have reached 20-40% in select contexts, paving the way for ex vivo gene therapies in hematopoietic cells. These developments underscore HR's role in transitioning from research tools to viable treatments for monogenic disorders.

Protein Engineering and Cancer Therapy

Homologous recombination (HR) plays a pivotal role in through techniques that mimic natural to evolve proteins with enhanced properties. One seminal method is , introduced in 1994, which involves random fragmentation of related genes followed by PCR-mediated reassembly to generate chimeric variants. This process leverages to facilitate recombination, enabling the rapid creation of diverse libraries for . For instance, DNA shuffling has been applied to evolve enzymes with improved catalytic efficiency, thermostability, and substrate specificity, accelerating the development of industrial biocatalysts beyond what random alone can achieve. In , HR-based recombination of homologous genes allows the combination of beneficial mutations from multiple parent sequences, often yielding superior variants. A notable example is the optimization of (GFP), where recursive of synthetic GFP variants with adjusted codon usage produced brighter and more stable fluorophores suitable for cellular imaging. This approach has been extended to engineer a range of proteins, including antibodies and metabolic enzymes, by recombining homologs from diverse species to explore functional diversity while maintaining structural integrity. Such HR-driven methods have become foundational in , enabling the tailoring of proteins for therapeutic and industrial applications without relying on structural predictions. In cancer therapy, defects in HR pathways, particularly in BRCA1/2-mutated tumors, create vulnerabilities that can be exploited through synthetic lethality. HR deficiency impairs the repair of double-strand breaks (DSBs), making cells reliant on alternative pathways like base excision repair, which poly(ADP-ribose) polymerase (PARP) enzymes facilitate. PARP inhibitors, such as olaparib, trap PARP on DNA and prevent single-strand break repair, leading to lethal DSB accumulation in HR-deficient cells while sparing normal cells with intact HR. Olaparib was the first PARP inhibitor approved for BRCA-mutated ovarian cancer, demonstrating significant progression-free survival benefits in clinical trials, with response rates up to 41% in germline BRCA carriers. This strategy has expanded to other HR-deficient tumors, including prostate and breast cancers, guided by genomic profiling for BRCAness phenotypes. HR defects also sensitize cancers to DSB-inducing agents like , a platinum-based chemotherapeutic that forms intrastrand crosslinks converted to DSBs during replication; in HR-proficient cells, these are repaired, but HR-deficient cells undergo catastrophic genomic instability. Combining with enhances this , as seen in preclinical models of BRCA-mutated where the duo induced greater tumor regression than either alone, due to compounded replication stress and unrepaired lesions. Emerging applications leverage HR for advanced immunotherapies, such as engineering chimeric antigen receptor () T cells via to precisely integrate CAR transgenes into safe genomic loci like , reducing tonic signaling and improving persistence. Recent advances in 2025 have refined HR-mediated CAR-T design to modulate surface density, enhancing efficacy against solid tumors while minimizing exhaustion.

References

  1. [1]
  2. [2]
    Homologous recombination and its regulation - PMC
    In this review, we focus on homologous recombination (HR), a mechanism that repairs a variety of DNA lesions, including double-strand DNA breaks (DSBs) ...
  3. [3]
    Mechanism and significance of chromosome damage repair by ...
    Aug 5, 2020 · Homologous recombination (HR) is a major, conserved pathway of chromosome damage repair. It not only fulfills key functions in the removal of deleterious ...
  4. [4]
    Chromosome architecture and homologous recombination in meiosis
    Homologous recombination during meiosis underlies biological diversity and ensures proper chromosome segregation during the first division to create haploid ...
  5. [5]
    Homologous recombination as a mechanism for genome ...
    HR is classically thought to be a mechanism for promoting genetic diversity. For example, in a diploid cell, meiotic recombination switches allele combinations ...
  6. [6]
    [PDF] Sturtevant, AH 1913. The linear arrangement of six sex-linked ...
    The idea that individual genes occupy regular positions on chromosomes was one of the great insights of early genetics, and the very first genetic map was ...
  7. [7]
    The Significance of Pneumococcal Types
    JANUARY, 1928. No. 2. THE SIGNIFICANCE OF PNEUMOCOCCAL TYPES. BY FRED. GRIFFITH, M.B.. (A Medical Officer of the Ministry of Health.) (From the Ministry's ...
  8. [8]
    [PDF] STUDIES ON THE CHEMICAL NATURE OF THE SUBSTANCE ...
    The task of Avery, Colin MacLeod and Maclyn McCarty was to purify this transforming substance--this genetic needle within the bacterial haystack. “The crude ...
  9. [9]
    [PDF] A correlation of cytological and genetical crossing-over in Zea mays.
    Such studies were first carried out on corn by Harriet B. Creighton and Barbara McClintock (this paper) and on Drosophila by Curt Stern.4. Both studies appeared ...
  10. [10]
    Conjugation and genetic recombination in Escherichia coli K-12
    Conjugation and genetic recombination in Escherichia coli K ... 1956:21:141-62. doi: 10.1101/sqb.1956.021.01.012. Authors. E L WOLLMAN, F JACOB ...Missing: bacterial studies original paper
  11. [11]
    A mechanism for gene conversion in fungi | Genetics Research
    Apr 14, 2009 · A mechanism for gene conversion is proposed which overcomes many of the difficulties that any copy choice model encounters.
  12. [12]
    The Nobel Prize in Chemistry 2015 - NobelPrize.org
    Prize share: 1/3. The Nobel Prize in Chemistry 2015 was awarded jointly to Tomas Lindahl, Paul Modrich and Aziz Sancar "for mechanistic studies of DNA repair".
  13. [13]
    Phylogenetic Ubiquity and Shuffling of the Bacterial RecBCD and ...
    Homologous recombination is highly conserved among viruses, bacteria, archaea, and eukaryotes. There are three recognizable stages (for reviews, see references ...<|separator|>
  14. [14]
    Origins and evolution of the recA/RAD51 gene family - PNAS
    Jul 5, 2006 · A major DSB repair pathway is homologous recombination, which is also critical for meiosis and generation of genetic diversity. Among the best ...Missing: definition | Show results with:definition
  15. [15]
    Mechanisms of horizontal gene transfer and DNA recombination
    Incoming DNA with significant similarity to the recipient genome can integrate by homologous recombination.
  16. [16]
    DNA repair by nonhomologous end joining and homologous ... - NIH
    The two major pathways for repair of DSBs are nonhomologous end joining (NHEJ) and homologous recombination (HR). NHEJ is an intrinsically error-prone pathway ...
  17. [17]
    Homologous recombination in DNA repair and DNA damage tolerance
    The DSB is processed to form a 3′-OH ending single-stranded tail (step 5a) and to initiate DNA strand invasion (step 6a). The replication fork is restored ...
  18. [18]
  19. [19]
    Repair of Strand Breaks by Homologous Recombination - PMC
    STEPS OF DSB REPAIR BY HR. The defining step of HR is the invasion of 3′ single-stranded DNA into a homologous duplex (Figs. 2 and 3). Single ...
  20. [20]
  21. [21]
    [PDF] lecture 5: linkage and genetic mapping - UC Berkeley MCB
    Recombination frequency = # recombinants/total progeny x 100. Experimental recombination frequencies between two genes are never greater than 50%. Recombinants ...
  22. [22]
    How RecBCD Enzyme and Chi Promote DNA Break Repair and ...
    I present evidence from both genetics and biochemistry indicating that for recombination and DNA break repair in Escherichia coli, Chi sites regulate the RecBCD ...
  23. [23]
    RecBCD Enzyme and the Repair of Double-Stranded DNA Breaks
    The RecBCD enzyme of Escherichia coli is a helicase-nuclease that initiates the repair of double-stranded DNA breaks by homologous recombination.
  24. [24]
    None
    ### Summary of the RecF Pathway
  25. [25]
  26. [26]
    The Role of the RecFOR Complex in Genome Stability - PMC
    Jun 6, 2025 · In contrast, in Mycobacteria, the RecFOR system has a central role in homology-dependent DNA repair, and RecR is essential for all three ...
  27. [27]
    sbcB15 and ΔsbcB Mutations Activate Two Types of RecF ...
    Escherichia coli cells with mutations in recBC genes are defective for the main RecBCD pathway of recombination and have severe reductions in conjugational ...<|control11|><|separator|>
  28. [28]
    Involvement of recF, recO, and recR Genes in UV-Radiation ...
    The recF, recO, and recR genes were originally identified as those affecting the RecF pathway of recombination in Escherichia coli cells.
  29. [29]
    Sending out an SOS - the bacterial DNA damage response - PMC
    It was found that in E. coli the recF pathway proteins, such as RecF, RecO, and RecR, are necessary to restore replication after UV radiation-induced damage. ...
  30. [30]
    recF and recR are required for the resumption of replication at DNA ...
    In Escherichia coli, recombination is classically thought to occur through one of two pathways termed the recBC (major) pathway and the recF (minor) pathway (1, ...
  31. [31]
    The Roles of Bacterial DNA Double-Strand Break Repair Proteins in ...
    Apr 14, 2020 · In this review, we analyze how bacterial DNA replication is perturbed in DSB repair mutant strains and explore the consequences of these perturbations.Recombinational Repair... · Recbcd Suppressors · Dna Amplification Of The...
  32. [32]
    Homologous recombination in DNA repair and DNA damage tolerance
    Jan 1, 2008 · This review focuses on the mechanism ... Homologous recombination: the core mechanism of Rad51 filament formation and DNA strand invasion.
  33. [33]
    The RAD51 recombinase protects mitotic chromatin in human cells
    Sep 10, 2021 · In this study, we show that RAD51 protects under-replicated DNA in mitotic human cells and, in this way, promotes mitotic DNA synthesis (MiDAS) and successful ...
  34. [34]
    Rescue of arrested replication forks by homologous recombination
    Arrested replication forks may be the target of nucleases, thereby providing a substrate for double-strand break repair enzyme. For example in bacteria, direct ...
  35. [35]
    Control of Meiotic Crossovers: From Double-Strand Break Formation ...
    In this review, we discuss the proteins involved in crossover formation, the process of their formation and designation, and the rules governing crossovers.
  36. [36]
    Initiation of Meiotic Homologous Recombination: Flexibility, Impact ...
    In this review, we outline our understanding of the control of ... From meiosis to postmeiotic events: Homologous recombination is obligatory but flexible.
  37. [37]
    The molecular machinery of meiotic recombination - Portland Press
    Meiotic recombination is an essential process, required at the organismal level to facilitate the proper segregation of homologous chromosomes, and at the ...
  38. [38]
    Chromosome architecture and homologous recombination in meiosis
    Homologous recombination during meiosis underlies biological diversity and ensures proper chromosome segregation during the first division to create haploid ...
  39. [39]
    Meiotic Crossover Patterning - Frontiers
    The synaptonemal complex imposes crossover interference and heterochiasmy in Arabidopsis. ... The synaptonemal complex has liquid crystalline properties and ...<|control11|><|separator|>
  40. [40]
    The choice in meiosis – defining the factors that influence crossover ...
    Feb 15, 2011 · Crossover interference: A mechanism that distributes meiotic DSBs and crossovers such that adjacent crossovers tend to occur further apart than ...
  41. [41]
    Meiotic Crossover Interference: Methods of Analysis and ...
    A DSB formed at random in the cluster ... The synaptonemal complex has liquid crystalline properties and spatially regulates meiotic recombination factors.
  42. [42]
    Roles for mismatch repair family proteins in promoting meiotic ...
    This review discusses how meiotic roles for Msh4-Msh5 and Mlh1-Mlh3 do not fit paradigms established for post-replicative MMR.
  43. [43]
    The mismatch repair and meiotic recombination endonuclease Mlh1 ...
    Mlh1-Mlh3 is recruited by Msh4-Msh5, which is then displaced by the Mlh1-Mlh3 polymer. The relative polarities of the two duplexes and the nicks resulting from ...
  44. [44]
    Meiotic Recombination: Mixing It Up in Plants - Annual Reviews
    Apr 29, 2018 · Meiotic recombination ensures the formation of bivalents between homologous chromosomes (homologs) and their subsequent proper segregation. It ...
  45. [45]
    Reduced meiotic crossovers and delayed prophase I progression in ...
    The role of AtMLH3 in CO formation. Evidence from budding yeast has confirmed that Msh4/Msh5 and Mlh1/Mlh3 function in the same recombination pathway (Hunter ...
  46. [46]
    Regulation of crossover frequency and distribution during meiotic ...
    The process of crossover recombination is tightly regulated and is initiated by the formation of programmed meiotic DNA double-strand breaks (DSBs).
  47. [47]
    The double-strand-break repair model for recombination - PubMed
    We propose a new mechanism for meiotic recombination, in which events are initiated by double-strand breaks that are enlarged to double-strand gaps.Missing: homologous | Show results with:homologous
  48. [48]
    Homologous recombination and the repair of DNA double-strand ...
    Homologous recombination enables the cell to access and copy intact DNA sequence information in trans, particularly to repair DNA damage.
  49. [49]
    Mechanism of Holliday junction resolution by the human GEN1 protein
    Holliday junction (HJ) resolution is essential for chromosome segregation at meiosis and the repair of stalled/collapsed replication forks in mitotic cells.
  50. [50]
    In vivo homologous recombination intermediates of yeast ... - PubMed
    Electron microscopic analysis revealed that these structures had pairs of equal length arms as required for homologous recombination intermediates and that ...Missing: double | Show results with:double
  51. [51]
  52. [52]
  53. [53]
    Hierarchy of nonhomologous end-joining, single-strand annealing ...
    Interestingly, Rad51 knockdown in CHO K1 cells resulted in a small yet robust increase in the frequency of SSA by 11% (Figure 5B; second bar), consistent ...
  54. [54]
    Break-induced replication: A review and an example in budding yeast
    Break-induced replication (BIR) is a nonreciprocal recombination-dependent replication process that is an effective mechanism to repair a broken chromosome.
  55. [55]
    Break-induced replication: unraveling each step - PMC - NIH
    Studies in yeast have demonstrated that DNA copying during BIR can continue for ~100 kb and through the telomere [6–10]. BIR synthesis occurs within a migrating ...
  56. [56]
    Break-induced replication requires all essential DNA replication ...
    Break-induced replication (BIR) is an efficient homologous recombination (HR) pathway employed to repair a DNA double-strand break (DSB) when homology is ...Missing: seminal | Show results with:seminal
  57. [57]
    Break-Induced Replication and Genome Stability - MDPI
    BIR is a DSB repair mechanism capable of recovering collapsed replication forks that is known to produce genomic rearrangements and genetic mutations at high ...5.2. Dna Synthesis... · 6. Mutagenesis Associated... · 7. Chromosomal...Missing: seminal | Show results with:seminal
  58. [58]
    Break-Induced Replication Requires DNA Damage-Induced ...
    Oct 16, 2014 · Break-Induced Replication Requires DNA Damage-Induced Phosphorylation of Pif1 and Leads to Telomere Lengthening.
  59. [59]
    A Microhomology-Mediated Break-Induced Replication Model for ...
    Jan 30, 2009 · We derive a model of microhomology-mediated break-induced replication (MMBIR) for the origin of CNV and, ultimately, of LCRs.
  60. [60]
    Recombination Promoted by DNA Viruses: Phage λ to Herpes ...
    Jun 9, 2014 · In this review we compare and contrast recombina- tion mechanisms in λ and HSV and explore how each virus utilizes recombination during its life.
  61. [61]
    Recombination in Positive-Strand RNA Viruses - Frontiers
    May 17, 2022 · Recombination occurs when two or more viruses co-infect a single cell and exchange segments, often giving rise to viable hybrid progeny. To ...
  62. [62]
    Why do RNA viruses recombine? | Nature Reviews Microbiology
    Jul 4, 2011 · The process of recombination that takes place in RNA viruses corresponds to the formation of chimeric molecules from parental genomes of mixed origin.
  63. [63]
    Homologous recombination is an intrinsic defense against antiviral ...
    Sep 12, 2018 · RNA interference (RNAi) is the major antiviral defense mechanism of plants and invertebrates, rendering the capacity to evade it a defining ...
  64. [64]
    Rad51 and Rad52 Are Involved in Homologous Recombination of ...
    Role of Rad51 and Rad52 in HSV-1 recombination. Existing results suggest a modest role for Rad51 during undisturbed DNA replication of the HSV-1 genome.
  65. [65]
    Small Genomes, Big Disruptions: Parvoviruses and the DNA ... - NIH
    Mar 29, 2025 · ATR signaling and RAD51 recruitment leading to homologous recombination are collectively inhibited by NS1. These interactions highlight how ...
  66. [66]
    Recombination in viruses: Mechanisms, methods of study, and ...
    Here we review three important aspects of viral recombination: (i) molecular mechanisms of model DNA- and RNA-viruses, (ii) methods for detection, ...
  67. [67]
    Herpes simplex virus 1 ICP8 mutant lacking annealing activity is ...
    Dec 31, 2018 · The herpes simplex virus (HSV) single-strand DNA binding protein (SSB), ICP8, is the central player in all stages of DNA replication. ICP8 is a ...
  68. [68]
    Copy-choice recombination by reverse transcriptases - PNAS
    Strand transfer occurring during reverse transcription of the region of homology between these two RNAs leads to the formation of molecules carrying the ...
  69. [69]
    Tracking HIV-1 recombination to resolve its contribution to ... - Nature
    May 15, 2018 · High-frequency illegitimate strand transfers of nascent DNA fragments during reverse transcription result in defective retrovirus genomes. J ...
  70. [70]
    Recombinant SARS-CoV-2 Delta/Omicron BA.5 emerging in an ...
    Oct 28, 2024 · In coronaviruses, these events are primarily driven by the RNA-dependent RNA polymerase, which can switch between viral templates during genome ...
  71. [71]
    Systematic detection of co-infection and intra-host recombination in ...
    Jan 15, 2024 · Here we present a comprehensive analysis of more than 2 million SARS-CoV-2 raw read datasets submitted to the European COVID-19 Data Portal to identify co- ...
  72. [72]
    The evolution of SARS-CoV-2 | Nature Reviews Microbiology
    Apr 5, 2023 · This Review explores the mechanisms that generate genetic variation in SARS-CoV-2, underlying the within-host and population-level processes that underpin ...
  73. [73]
    Defective viral genomes are key drivers of the virus–host interaction
    Jun 3, 2019 · Replication-driven template-switching. RNA recombination is a major driver of deletion DVG formation. A predominant model proposes that ...
  74. [74]
    Unresolved recombination intermediates lead to ultra-fine anaphase ...
    Unresolved recombination intermediates lead to ultra-fine anaphase bridges, chromosome breaks and aberrations. Nat Cell Biol. 2018 Jan;20(1):92-103.
  75. [75]
    Loss of heterozygosity preferentially occurs in early replicating ...
    Jun 21, 2013 · Abstract. Erroneous repair of DNA double-strand breaks by homologous recombination (HR) leads to loss of heterozygosity (LOH).
  76. [76]
    Faithful after break-up: suppression of chromosomal translocations
    Substantial evidence links HR, in particular, BIR and SSA, to DSB-induced ... chromosomal rearrangements including chromosomal translocations by SSA.
  77. [77]
    Break‐induced replication plays a prominent role in long‐range ...
    Oct 1, 2019 · We demonstrate that break‐induced replication (BIR) is used predominantly over SSA in mammalian cells for mediating RMD, especially when repeats are far apart.
  78. [78]
    RAD52: Paradigm of Synthetic Lethality and New Developments
    Nov 23, 2021 · Although it is critical for most DNA repair and recombination events in yeast, knockouts of mammalian RAD52 lack any discernable phenotypes.
  79. [79]
    Rad52 inactivation is synthetically lethal with BRCA2 deficiency
    We show that loss of Rad52 function is synthetically lethal with breast cancer 2, early onset (BRCA2) deficiency, whereas there was no impact on cell growth ...Rad52 Inactivation Is... · Results · Depletion Of Rad52 By Shrna...Missing: hyper- | Show results with:hyper-
  80. [80]
    Aging impairs double‐strand break repair by homologous ... - NIH
    Dec 21, 2016 · Aging is characterized by genome instability, which contributes to cancer formation and cell lethality leading to organismal decline.
  81. [81]
    Aging impairs double‐strand break repair by homologous ...
    Dec 21, 2016 · Aging is characterized by genome instability, which contributes to cancer formation and cell lethality leading to organismal decline.
  82. [82]
    Replication Stress Induces Micronuclei Comprising of Aggregated ...
    Apr 15, 2011 · Elevation of MN is commonly observed in cells bearing intrinsic genomic instability and in cells exposed to genotoxic agents. Compared to assays ...
  83. [83]
    Interplay between Fanconi anemia and homologous recombination ...
    In this review, we highlight the clinical features ... Fanconi anemia proteins FANCD2 and FANCJ interact and regulate each other's chromatin localization.Missing: paper | Show results with:paper
  84. [84]
    Human Fanconi anemia monoubiquitination pathway promotes ...
    We find that both the upstream (FANCA and FANCG) and downstream (FANCD2) FA pathway components promote homology-directed repair of chromosomal double-strand ...Missing: review | Show results with:review
  85. [85]
    Review Homologous recombination deficiency and ovarian cancer
    BRCA1 and BRCA2 proteins are essential for high-fidelity repair of double-strand breaks of DNA through the homologous recombination repair (HRR) pathway.
  86. [86]
    Homologous Recombination Deficiency in Ovarian, Breast ...
    Findings showed that BRCA1 and 2 inactivation frequently led to higher HRD score in ovarian and breast cancers. This high HRD score has positive prognostic ...
  87. [87]
    Pan-cancer landscape of homologous recombination deficiency
    Nov 4, 2020 · We identify inactivation of BRCA1, BRCA2, RAD51C, and PALB2 as the most frequent genetic cause of HRD pan-cancer in both primary and metastatic ...
  88. [88]
    Functions of BLM Helicase in Cells: Is It Acting Like a Double-Edged ...
    Mar 12, 2021 · Genetic mutations in the BLM gene cause a rare, autosomal recessive disorder, Bloom syndrome (BS). BS is a monogenic disease characterized by ...
  89. [89]
    and anti-recombination activities of the Bloom's syndrome helicase
    Nov 14, 2007 · BLM plays a role in homologous recombination; however, its exact function remains controversial. Mutations in the BLM cause hyperrecombination ...Missing: hyper- | Show results with:hyper-
  90. [90]
    The molecular control of meiotic double-strand break (DSB ...
    Feb 12, 2021 · We further reviewed the mutations of DSB formation genes in association with human infertility and also proposed the future directions and ...
  91. [91]
    Genetic diagnosis of subfertility: the impact of meiosis and maternal ...
    In this review, we specifically focus on genes involved in meiosis and maternal-effect processes, which are of critical importance for reproduction and initial ...
  92. [92]
    Synthetic lethality of PARP inhibition in cancers lacking BRCA1 and ...
    Inhibition of PARP was subsequently shown to cause selective cell death in ATM deficient cancer cell lines due to their impaired HR by our group where siRNA ...
  93. [93]
    The underlying mechanism for the PARP and BRCA synthetic lethality
    ... PARP inhibitors mediate selective killing of HR defective cells. Future ... Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) ...
  94. [94]
    The structure of the E. coli recA protein monomer and polymer - Nature
    Jan 23, 1992 · The crystal structure of the recA protein from Escherichia coli at 2.3-Å resolution reveals a major domain that binds ADP and probably single- and double- ...
  95. [95]
    RecA: Regulation and Mechanism of a Molecular Search Engine - NIH
    Homologous recombination maintains genomic integrity by repairing broken chromosomes. The broken chromosome is partially resected to produce single-stranded DNA ...
  96. [96]
    Mechanism of RecA-mediated homologous recombination revisited ...
    Lindsley JE, Cox M (1990) Assembly and disassembly of RecA protein filaments occur at opposite filament ends. Relationship to DNA strand exchange. J Biol ...
  97. [97]
    Interactions between human BRCA2 protein and the meiosis ...
    In addition to RAD51, meiotic recombination requires the functions of DMC1, a meiosis‐specific paralog of RAD51 (Bishop et al, 1992; Yoshida et al, 1998). Like ...
  98. [98]
    Crystal Structure of Archaeal Recombinase RadA: A Snapshot of Its ...
    Rad51, DMC1, and RadA possess a somewhat conserved N-terminal domain, while RecA has a similar domain at its C terminus (Yang et al., 2001a). At the sequence ...
  99. [99]
    The Evolution of Meiosis From Mitosis - PMC - PubMed Central - NIH
    While meiosis almost certainly evolved from mitosis, it has not one but four novel steps: the pairing of homologous chromosomes, the occurrence of extensive ...
  100. [100]
    Spo11 and the Formation of DNA Double-Strand Breaks in Meiosis
    Spo11 catalyzes DNA double-strand break formation in meiosis, acting via a topoisomerase-like reaction, and is related to archaeal topoisomerase VI.
  101. [101]
    Meiosis in budding yeast - PMC - PubMed Central
    ... spo11 mutant displays chromosome individualization defects similar to red1 and hop1 mutants (Mao-Draayer et al. 1996; Smith and Roeder 1997; Klein et al ...
  102. [102]
    A conserved filamentous assembly underlies the structure of ... - eLife
    Jan 18, 2019 · In budding yeast, the chromosome axis is made up of the HORMAD protein Hop1, its binding partner Red1, and cohesin complexes containing the ...
  103. [103]
    Genetic Interactions between Hop1, Red1 and Mek1 Suggest ... - NIH
    Genetic and cytological data suggest that three meiosis-specific genes, HOP1, RED1 and MEK1, are involved in axial element formation in the yeast Saccharomyces ...Missing: seminal | Show results with:seminal
  104. [104]
    Reduced dosage of the chromosome axis factor Red1 selectively ...
    Our results indicate separable roles for Red1 in building the structural axis of meiotic chromosomes and mounting a sustained recombination checkpoint response.
  105. [105]
    Msh4 and Msh5 Function in SC-Independent Chiasma Formation ...
    Here we show that the ZMM proteins Msh4 and Msh5 are required for normal chiasma formation, and we propose that they have a pro-CO function outside a canonical ...
  106. [106]
    Coupling crossover and synaptonemal complex in meiosis
    (Left panel) The ZMM proteins (Zip4, Zip2, Spo16, Msh4, Msh5, Zip3, Zip1, and Mer3) channel DSB repair toward the homolog and stabilize the D-loop intermediate.
  107. [107]
    The synaptonemal complex protein, Zip1, promotes the segregation ...
    Dec 22, 2009 · During meiosis, Msh4 forms a heterodimer with Msh5 (25), which recognizes double Holliday junctions in vitro (26). Together with Zip3, Zip1, ...Missing: class | Show results with:class
  108. [108]
    Evolutionary Origin of Recombination during Meiosis | BioScience
    Recent evidence indicates that meiosis arose very early in eukaryotic evolution, which suggests that essential features of meiosis were already present in the ...
  109. [109]
    CRISPR-Cas9-mediated homology-directed repair for precise gene ...
    CRISPR-Cas9-mediated homology-directed repair (HDR) is a versatile platform for creating precise site-specific DNA insertions, deletions, and substitutions.
  110. [110]
    CRISPR 101: Homology Directed Repair - Addgene Blog
    Jan 26, 2023 · HDR donors can contain insertable sequences or specific mutations, along with homologous DNA to the target sequence to be modified. In general, ...
  111. [111]
    ZFN, TALEN and CRISPR/Cas-based methods for genome ...
    ZFNs and TALENs enable a broad range of genetic modifications by inducing DNA double-strand breaks that stimulate error-prone non-homologous end joining.
  112. [112]
    Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR ...
    Oct 1, 2014 · Gene targeting by homologous recombination in mouse zygotes mediated by zinc-finger nucleases. Proc Natl Acad Sci U S A. 2010;107(34):15022 ...
  113. [113]
    Efficient CRISPR/Cas9-mediated gene editing in mammalian cells ...
    Usually, the HDR-based knock-in efficiency is pretty low (1–10 % of modified alleles) even in the enriched cells screened by the NHEJ- and SSA-based reporters.
  114. [114]
    Modulation of cell cycle increases CRISPR-mediated homology ...
    Nov 25, 2023 · Small molecules that induce cell cycle synchronization in S and G2/M phases promote CRISPR/Cas9-mediated HDR efficiency in animal cells and embryos.
  115. [115]
    CRISPR-Cas9-mediated homology-directed repair for precise gene ...
    Here, we review CRISPR-Cas9-mediated HDR, with a focus on methodologies for boosting HDR efficiency, and applications of precise editing via HDR. First, we ...
  116. [116]
    Recent advances in therapeutic gene-editing technologies
    Jun 4, 2025 · This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.<|separator|>
  117. [117]
    Rapid evolution of a protein in vitro by DNA shuffling - Nature
    Aug 4, 1994 · DNA SHUFFLING is a method for in vitro homologous recombination of pools of selected mutant genes by random fragmentation and polymerase chain reaction (PCR) ...
  118. [118]
    Improved Green Fluorescent Protein by Molecular Evolution Using ...
    Mar 1, 1996 · We constructed a synthetic GFP gene with improved codon usage and performed recursive cycles of DNA shuffling followed by screening for the brightest E. coli ...Missing: directed | Show results with:directed
  119. [119]
    Targeting the DNA repair defect in BRCA mutant cells as ... - PubMed
    We show here that BRCA1 or BRCA2 dysfunction unexpectedly and profoundly sensitizes cells to the inhibition of PARP enzymatic activity.
  120. [120]
    Inhibition of Poly(ADP-Ribose) Polymerase in Tumors from BRCA ...
    Jun 24, 2009 · Such “synthetic lethality” occurs when there is a potent and lethal synergy between two otherwise nonlethal events: in this case, a highly ...
  121. [121]
    Synthetic lethality: the road to novel therapies for breast cancer in
    Therefore, identification of additional drugs that can effectively treat HR-deficient cancers by exploiting synthetic lethal gene interactions is essential (Fig ...
  122. [122]
    Enhancing synthetic lethality of PARP-inhibitor and cisplatin in ... - NIH
    Inactive HR can be due to mutations in BRCA1 or BRCA2, which may result in potentially lethal accumulation of DNA double strand breaks (DSBs). HR-deficient ( ...
  123. [123]
    Homology-Directed Recombination for Enhanced Engineering ... - NIH
    Here, we demonstrate the efficiency and effectiveness of this approach to generate gene-targeted CAR T cells with concurrent disruption in these clinically ...Missing: 2024 2025<|control11|><|separator|>
  124. [124]
    Tailoring CAR surface density and dynamics to improve CAR-T cell ...
    Apr 29, 2025 · CAR expression can also be regulated by targeting a specific endogenous gene and inserting it via homologous recombination into its locus, ...