fbpx
Wikipedia

Homologous recombination

Homologous recombination is a type of genetic recombination in which genetic information is exchanged between two similar or identical molecules of double-stranded or single-stranded nucleic acids (usually DNA as in cellular organisms but may be also RNA in viruses).

Figure 1. During meiosis, homologous recombination can produce new combinations of genes as shown here between similar but not identical copies of human chromosome 1.

Homologous recombination is widely used by cells to accurately repair harmful DNA breaks that occur on both strands of DNA, known as double-strand breaks (DSB), in a process called homologous recombinational repair (HRR).[1]

Homologous recombination also produces new combinations of DNA sequences during meiosis, the process by which eukaryotes make gamete cells, like sperm and egg cells in animals. These new combinations of DNA represent genetic variation in offspring, which in turn enables populations to adapt during the course of evolution.[2]

Homologous recombination is also used in horizontal gene transfer to exchange genetic material between different strains and species of bacteria and viruses. Horizontal gene transfer is the primary mechanism for the spread of antibiotic resistance in bacteria.

Although homologous recombination varies widely among different organisms and cell types, for double-stranded DNA (dsDNA) most forms involve the same basic steps. After a double-strand break occurs, sections of DNA around the 5' ends of the break are cut away in a process called resection. In the strand invasion step that follows, an overhanging 3' end of the broken DNA molecule then "invades" a similar or identical DNA molecule that is not broken. After strand invasion, the further sequence of events may follow either of two main pathways discussed below (see Models); the DSBR (double-strand break repair) pathway or the SDSA (synthesis-dependent strand annealing) pathway. Homologous recombination that occurs during DNA repair tends to result in non-crossover products, in effect restoring the damaged DNA molecule as it existed before the double-strand break.

Homologous recombination is conserved across all three domains of life as well as DNA and RNA viruses, suggesting that it is a nearly universal biological mechanism. The discovery of genes for homologous recombination in protists—a diverse group of eukaryotic microorganisms—has been interpreted as evidence that homologous recombination emerged early in the evolution of eukaryotes. Since their dysfunction has been strongly associated with increased susceptibility to several types of cancer, the proteins that facilitate homologous recombination are topics of active research. Homologous recombination is also used in gene targeting, a technique for introducing genetic changes into target organisms. For their development of this technique, Mario Capecchi, Martin Evans and Oliver Smithies were awarded the 2007 Nobel Prize for Physiology or Medicine; Capecchi[3] and Smithies[4] independently discovered applications to mouse embryonic stem cells, however the highly conserved mechanisms underlying the DSB repair model, including uniform homologous integration of transformed DNA (gene therapy), were first shown in plasmid experiments by Orr-Weaver, Szostak and Rothstein.[5][6][7] Researching the plasmid-induced DSB, using γ-irradiation[8] in the 1970s-1980s, led to later experiments using endonucleases (e.g. I-SceI) to cut chromosomes for genetic engineering of mammalian cells, where nonhomologous recombination is more frequent than in yeast.[9]

History and discovery edit

 
Figure 2. An early illustration of crossing over from Thomas Hunt Morgan

In the early 1900s, William Bateson and Reginald Punnett found an exception to one of the principles of inheritance originally described by Gregor Mendel in the 1860s. In contrast to Mendel's notion that traits are independently assorted when passed from parent to child—for example that a cat's hair color and its tail length are inherited independent of each other—Bateson and Punnett showed that certain genes associated with physical traits can be inherited together, or genetically linked.[10][11] In 1911, after observing that linked traits could on occasion be inherited separately, Thomas Hunt Morgan suggested that "crossovers" can occur between linked genes,[12] where one of the linked genes physically crosses over to a different chromosome. Two decades later, Barbara McClintock and Harriet Creighton demonstrated that chromosomal crossover occurs during meiosis,[13][14] the process of cell division by which sperm and egg cells are made. Within the same year as McClintock's discovery, Curt Stern showed that crossing over—later called "recombination"—could also occur in somatic cells like white blood cells and skin cells that divide through mitosis.[13][15]

In 1947, the microbiologist Joshua Lederberg showed that bacteria—which had been assumed to reproduce only asexually through binary fission—are capable of genetic recombination, which is more similar to sexual reproduction. This work established E. coli as a model organism in genetics,[16] and helped Lederberg win the 1958 Nobel Prize in Physiology or Medicine.[17] Building on studies in fungi, in 1964 Robin Holliday proposed a model for recombination in meiosis which introduced key details of how the process can work, including the exchange of material between chromosomes through Holliday junctions.[18] In 1983, Jack Szostak and colleagues presented a model now known as the DSBR pathway, which accounted for observations not explained by the Holliday model.[18][7] During the next decade, experiments in Drosophila, budding yeast and mammalian cells led to the emergence of other models of homologous recombination, called SDSA pathways, which do not always rely on Holliday junctions.[18]

Much of the later work identifying proteins involved in the process and determining their mechanisms has been performed by a number of individuals including James Haber, Patrick Sung, Stephen Kowalczykowski, and others.

In eukaryotes edit

Homologous recombination (HR) is essential to cell division in eukaryotes like plants, animals, fungi and protists. Homologous recombination repairs double-strand breaks in DNA caused by ionizing radiation or DNA-damaging chemicals.[19] Left unrepaired, these double-strand breaks can cause large-scale rearrangement of chromosomes in somatic cells,[20] which can in turn lead to cancer.[21]

In addition to repairing DNA, homologous recombination also helps produce genetic diversity when cells divide in meiosis to become specialized gamete cells—sperm or egg cells in animals, pollen or ovules in plants, and spores in fungi. It does so by facilitating chromosomal crossover, in which regions of similar but not identical DNA are exchanged between homologous chromosomes.[22][23] This creates new, possibly beneficial combinations of genes, which can give offspring an evolutionary advantage.[24] Chromosomal crossover often begins when a protein called Spo11 makes a targeted double-strand break in DNA.[25] These sites are non-randomly located on the chromosomes; usually in intergenic promoter regions and preferentially in GC-rich domains[26] These double-strand break sites often occur at recombination hotspots, regions in chromosomes that are about 1,000–2,000 base pairs in length and have high rates of recombination. The absence of a recombination hotspot between two genes on the same chromosome often means that those genes will be inherited by future generations in equal proportion. This represents linkage between the two genes greater than would be expected from genes that independently assort during meiosis.[27]

Timing within the mitotic cell cycle edit

 
Figure 3. Homologous recombination repair attempts occur in DNA before the cell enters mitosis (M phase shown) during the S and G2 phases of the cell cycle.

Double-strand breaks can be repaired through homologous recombination, polymerase theta-mediated end joining (TMEJ) or through non-homologous end joining (NHEJ).[28] NHEJ is a DNA repair mechanism which, unlike homologous recombination, does not require a long homologous sequence to guide repair. Whether homologous recombination or NHEJ is used to repair double-strand breaks is largely determined by the phase of cell cycle. Homologous recombination repairs DNA before the cell enters mitosis (M phase). It occurs during and shortly after DNA replication, in the S and G2 phases of the cell cycle, when sister chromatids are more easily available.[29] Compared to homologous chromosomes, which are similar to another chromosome but often have different alleles, sister chromatids are an ideal template for homologous recombination because they are an identical copy of a given chromosome. When no homologous template is available or when the template cannot be accessed due to a defect in homologous recombination, the break is repaired via TMEJ in the S and G2 phases of the cell cycle. In contrast to homologous recombination and TMEJ, NHEJ is predominant in the G1 phase of the cell cycle, when the cell is growing but not yet ready to divide. It occurs less frequently after the G1 phase, but maintains at least some activity throughout the cell cycle. The mechanisms that regulate homologous recombination and NHEJ throughout the cell cycle vary widely between species.[30]

Cyclin-dependent kinases (CDKs), which modify the activity of other proteins by adding phosphate groups to (that is, phosphorylating) them, are important regulators of homologous recombination in eukaryotes.[30] When DNA replication begins in budding yeast, the cyclin-dependent kinase Cdc28 begins homologous recombination by phosphorylating the Sae2 protein.[31] After being so activated by the addition of a phosphate, Sae2 causes a clean cut to be made near a double-strand break in DNA. It is unclear if the endonuclease responsible for this cut is Sae2 itself or another protein, Mre11.[32] This allows a protein complex including Mre11, known as the MRX complex, to bind to DNA, and begins a series of protein-driven reactions that exchange material between two DNA molecules.[33]

The role of chromatin edit

The packaging of eukaryotic DNA into chromatin presents a barrier to all DNA-based processes that require recruitment of enzymes to their sites of action. To allow homologous recombination (HR) DNA repair, the chromatin must be remodeled. In eukaryotes, ATP dependent chromatin remodeling complexes and histone-modifying enzymes are two predominant factors employed to accomplish this remodeling process.[34]

Chromatin relaxation occurs rapidly at the site of a DNA damage.[35] In one of the earliest steps, the stress-activated protein kinase, c-Jun N-terminal kinase (JNK), phosphorylates SIRT6 on serine 10 in response to double-strand breaks or other DNA damage.[36] This post-translational modification facilitates the mobilization of SIRT6 to DNA damage sites, and is required for efficient recruitment of poly (ADP-ribose) polymerase 1 (PARP1) to DNA break sites and for efficient repair of DSBs.[36] PARP1 protein starts to appear at DNA damage sites in less than a second, with half maximum accumulation within 1.6 seconds after the damage occurs.[37] Next the chromatin remodeler Alc1 quickly attaches to the product of PARP1 action, a poly-ADP ribose chain, and Alc1 completes arrival at the DNA damage within 10 seconds of the occurrence of the damage.[35] About half of the maximum chromatin relaxation, presumably due to action of Alc1, occurs by 10 seconds.[35] This then allows recruitment of the DNA repair enzyme MRE11, to initiate DNA repair, within 13 seconds.[37]

γH2AX, the phosphorylated form of H2AX is also involved in the early steps leading to chromatin decondensation after DNA double-strand breaks. The histone variant H2AX constitutes about 10% of the H2A histones in human chromatin.[38] γH2AX (H2AX phosphorylated on serine 139) can be detected as soon as 20 seconds after irradiation of cells (with DNA double-strand break formation), and half maximum accumulation of γH2AX occurs in one minute.[38] The extent of chromatin with phosphorylated γH2AX is about two million base pairs at the site of a DNA double-strand break.[38] γH2AX does not, itself, cause chromatin decondensation, but within 30 seconds of irradiation, RNF8 protein can be detected in association with γH2AX.[39] RNF8 mediates extensive chromatin decondensation, through its subsequent interaction with CHD4,[40] a component of the nucleosome remodeling and deacetylase complex NuRD.

After undergoing relaxation subsequent to DNA damage, followed by DNA repair, chromatin recovers to a compaction state close to its pre-damage level after about 20 min.[35]

Homologous recombination during meiosis edit

In vertebrates the locations at which recombination occurs are determined by the binding locations of PRDM9, a protein which recognizes a specific sequence motif by its zinc finger array.[41] At these sites, another protein, SPO11 catalyses recombination-initiating double strand breaks (DSBs), a subset of which are repaired by recombination with the homologous chromosome. PRDM9 deposits both H3K4me3 and H3K36me3 histone methylation marks at the sites it binds, and this methyltransferase activity is essential for its role in DSB positioning. Following their formation, DSB sites are processed by resection, resulting in single-stranded DNA (ssDNA) that becomes decorated with DMC1. From mid-zygotene to early pachytene, as part of the recombinational repair process, DMC1 dissociates from the ssDNA and counts decrease until all breaks (except those on the XY chromosomes) are repaired at late pachytene. Several other proteins are involved in this process, including ZCWPW1, the [42] first protein directly positioned by PRDM9's dual histone marks. ZCWPW1 is important for homologous DSB repair, not positioning.

Models edit

 
Figure 4. Double-strand break repair models that act via homologous recombination

Two primary models for how homologous recombination repairs double-strand breaks in DNA are the double-strand break repair (DSBR) pathway (sometimes called the double Holliday junction model) and the synthesis-dependent strand annealing (SDSA) pathway.[43] The two pathways are similar in their first several steps. After a double-strand break occurs, the MRX complex (MRN complex in humans) binds to DNA on either side of the break. Next a resection takes place, in which DNA around the 5' ends of the break is cut back. This happens in two distinct steps: first the MRX complex recruits the Sae2 protein, and these two proteins trim back the 5' ends on either side of the break to create short 3' overhangs of single-strand DNA; in the second step, 5'→3' resection is continued by the Sgs1 helicase and the Exo1 and Dna2 nucleases. As a helicase, Sgs1 "unzips" the double-strand DNA, while the nuclease activity of Exo1 and Dna2 allows them to cut the single-stranded DNA produced by Sgs1.[31]

The RPA protein, which has high affinity for single-stranded DNA, then binds the 3' overhangs.[44] With the help of several other proteins that mediate the process, the Rad51 protein (and Dmc1, in meiosis) then forms a filament of nucleic acid and protein on the single strand of DNA coated with RPA. This nucleoprotein filament then begins searching for DNA sequences similar to that of the 3' overhang. After finding such a sequence, the single-stranded nucleoprotein filament moves into (invades) the similar or identical recipient DNA duplex in a process called strand invasion. In cells that divide through mitosis, the recipient DNA duplex is generally a sister chromatid, which is identical to the damaged DNA molecule and provides a template for repair. In meiosis, however, the recipient DNA tends to be from a similar but not necessarily identical homologous chromosome.[43] A displacement loop (D-loop) is formed during strand invasion between the invading 3' overhang strand and the homologous chromosome. After strand invasion, a DNA polymerase extends the end of the invading 3' strand by synthesizing new DNA. This changes the D-loop to a cross-shaped structure known as a Holliday junction. Following this, more DNA synthesis occurs on the invading strand (i.e., one of the original 3' overhangs), effectively restoring the strand on the homologous chromosome that was displaced during strand invasion.[43]

DSBR pathway edit

 
Figure 5. The DSBR and SDSA pathways follow the same initial steps, but diverge thereafter. The DSBR pathway most often results in chromosomal crossover (bottom left), while SDSA always ends with non-crossover products (bottom right).

After the stages of resection, strand invasion and DNA synthesis, the DSBR and SDSA pathways become distinct.[43] The DSBR pathway is unique in that the second 3' overhang (which was not involved in strand invasion) also forms a Holliday junction with the homologous chromosome. The double Holliday junctions are then converted into recombination products by nicking endonucleases, a type of restriction endonuclease which cuts only one DNA strand. The DSBR pathway commonly results in crossover, though it can sometimes result in non-crossover products; the ability of a broken DNA molecule to collect sequences from separated donor loci was shown in mitotic budding yeast using plasmids or endonuclease induction of chromosomal events.[45][46] Because of this tendency for chromosomal crossover, the DSBR pathway is a likely model of how crossover homologous recombination occurs during meiosis.[22]

Whether recombination in the DSBR pathway results in chromosomal crossover is determined by how the double Holliday junction is cut, or "resolved". Chromosomal crossover will occur if one Holliday junction is cut on the crossing strand and the other Holliday junction is cut on the non-crossing strand (in Figure 5, along the horizontal purple arrowheads at one Holliday junction and along the vertical orange arrowheads at the other). Alternatively, if the two Holliday junctions are cut on the crossing strands (along the horizontal purple arrowheads at both Holliday junctions in Figure 5), then chromosomes without crossover will be produced.[47]

SDSA pathway edit

Homologous recombination via the SDSA pathway occurs in cells that divide through mitosis and meiosis and results in non-crossover products. In this model, the invading 3' strand is extended along the recipient DNA duplex by a DNA polymerase, and is released as the Holliday junction between the donor and recipient DNA molecules slides in a process called branch migration. The newly synthesized 3' end of the invading strand is then able to anneal to the other 3' overhang in the damaged chromosome through complementary base pairing. After the strands anneal, a small flap of DNA can sometimes remain. Any such flaps are removed, and the SDSA pathway finishes with the resealing, also known as ligation, of any remaining single-stranded gaps.[48]

During mitosis, the major homologous recombination pathway for repairing DNA double-strand breaks appears to be the SDSA pathway (rather than the DSBR pathway).[49] The SDSA pathway produces non-crossover recombinants (Figure 5). During meiosis non-crossover recombinants also occur frequently and these appear to arise mainly by the SDSA pathway as well.[49][50] Non-crossover recombination events occurring during meiosis likely reflect instances of repair of DNA double-strand damages or other types of DNA damages.

SSA pathway edit

 
Figure 6. Recombination via the SSA pathway occurs between two repeat elements (purple) on the same DNA duplex, and results in deletions of genetic material. (Click to view animated diagram in Firefox, Chrome, Safari, or Opera web browsers.)

The single-strand annealing (SSA) pathway of homologous recombination repairs double-strand breaks between two repeat sequences. The SSA pathway is unique in that it does not require a separate similar or identical molecule of DNA, like the DSBR or SDSA pathways of homologous recombination. Instead, the SSA pathway only requires a single DNA duplex, and uses the repeat sequences as the identical sequences that homologous recombination needs for repair. The pathway is relatively simple in concept: after two strands of the same DNA duplex are cut back around the site of the double-strand break, the two resulting 3' overhangs then align and anneal to each other, restoring the DNA as a continuous duplex.[48][51]

As DNA around the double-strand break is cut back, the single-stranded 3' overhangs being produced are coated with the RPA protein, which prevents the 3' overhangs from sticking to themselves.[52] A protein called Rad52 then binds each of the repeat sequences on either side of the break, and aligns them to enable the two complementary repeat sequences to anneal.[52] After annealing is complete, leftover non-homologous flaps of the 3' overhangs are cut away by a set of nucleases, known as Rad1/Rad10, which are brought to the flaps by the Saw1 and Slx4 proteins.[52][53] New DNA synthesis fills in any gaps, and ligation restores the DNA duplex as two continuous strands.[54] The DNA sequence between the repeats is always lost, as is one of the two repeats. The SSA pathway is considered mutagenic since it results in such deletions of genetic material.[48]

BIR pathway edit

During DNA replication, double-strand breaks can sometimes be encountered at replication forks as DNA helicase unzips the template strand. These defects are repaired in the break-induced replication (BIR) pathway of homologous recombination. The precise molecular mechanisms of the BIR pathway remain unclear. Three proposed mechanisms have strand invasion as an initial step, but they differ in how they model the migration of the D-loop and later phases of recombination.[55]

The BIR pathway can also help to maintain the length of telomeres (regions of DNA at the end of eukaryotic chromosomes) in the absence of (or in cooperation with) telomerase. Without working copies of the enzyme telomerase, telomeres typically shorten with each cycle of mitosis, which eventually blocks cell division and leads to senescence. In budding yeast cells where telomerase has been inactivated through mutations, two types of "survivor" cells have been observed to avoid senescence longer than expected by elongating their telomeres through BIR pathways.[55]

Maintaining telomere length is critical for cell immortalization, a key feature of cancer. Most cancers maintain telomeres by upregulating telomerase. However, in several types of human cancer, a BIR-like pathway helps to sustain some tumors by acting as an alternative mechanism of telomere maintenance.[56] This fact has led scientists to investigate whether such recombination-based mechanisms of telomere maintenance could thwart anti-cancer drugs like telomerase inhibitors.[57]

In bacteria edit

 
Figure 7. Crystal structure of a RecA protein filament bound to DNA.[58] A 3' overhang is visible to the right of center.

Homologous recombination is a major DNA repair process in bacteria. It is also important for producing genetic diversity in bacterial populations, although the process differs substantially from meiotic recombination, which repairs DNA damages and brings about diversity in eukaryotic genomes. Homologous recombination has been most studied and is best understood for Escherichia coli.[59] Double-strand DNA breaks in bacteria are repaired by the RecBCD pathway of homologous recombination. Breaks that occur on only one of the two DNA strands, known as single-strand gaps, are thought to be repaired by the RecF pathway.[60] Both the RecBCD and RecF pathways include a series of reactions known as branch migration, in which single DNA strands are exchanged between two intercrossed molecules of duplex DNA, and resolution, in which those two intercrossed molecules of DNA are cut apart and restored to their normal double-stranded state.

RecBCD pathway edit

 
Figure 8A. Molecular model for the RecBCD pathway of recombination. This model is based on reactions of DNA and RecBCD with ATP in excess over Mg2+ ions. Step 1: RecBCD binds to a double-stranded DNA end. Step 2: RecBCD unwinds DNA. RecD is a fast helicase on the 5’-ended strand, and RecB is a slower helicase on the 3'-ended strand (that with an arrowhead) [ref 46 in current Wiki version]. This produces two single-stranded (ss) DNA tails and one ss loop. The loop and tails enlarge as RecBCD moves along the DNA. Step 3: The two tails anneal to produce a second ss DNA loop, and both loops move and grow. Step 4: Upon reaching the Chi hotspot sequence (5' GCTGGTGG 3'; red dot) RecBCD nicks the 3’-ended strand. Further unwinding produces a long 3'-ended ss tail with Chi near its end. Step 5: RecBCD loads RecA protein onto the Chi tail. At some undetermined point, the RecBCD subunits disassemble. Step 6: The RecA-ssDNA complex invades an intact homologous duplex DNA to produce a D-loop, which can be resolved into intact, recombinant DNA in two ways. Step 7: The D-loop is cut and anneals with the gap in the first DNA to produce a Holliday junction. Resolution of the Holliday junction (cutting, swapping of strands, and ligation) at the open arrowheads by some combination of RuvABC and RecG produces two recombinants of reciprocal type. Step 8: The 3' end of the Chi tail primes DNA synthesis, from which a replication fork can be generated. Resolution of the fork at the open arrowheads produces one recombinant (non-reciprocal) DNA, one parental-type DNA, and one DNA fragment.[61]
 
Figure 8B. Beginning of the RecBCD pathway. This model is based on reactions of DNA and RecBCD with Mg2+ ions in excess over ATP. Step 1: RecBCD binds to a DNA double strand break. Step 2: RecBCD initiates unwinding of the DNA duplex through ATP-dependent helicase activity. Step 3: RecBCD continues its unwinding and moves down the DNA duplex, cleaving the 3' strand much more frequently than the 5' strand. Step 4: RecBCD encounters a Chi sequence and stops digesting the 3' strand; cleavage of the 5' strand is significantly increased. Step 5: RecBCD loads RecA onto the 3' strand. Step 6: RecBCD unbinds from the DNA duplex, leaving a RecA nucleoprotein filament on the 3' tail.[62]

The RecBCD pathway is the main recombination pathway used in many bacteria to repair double-strand breaks in DNA, and the proteins are found in a broad array of bacteria.[63][64][65] These double-strand breaks can be caused by UV light and other radiation, as well as chemical mutagens. Double-strand breaks may also arise by DNA replication through a single-strand nick or gap. Such a situation causes what is known as a collapsed replication fork and is fixed by several pathways of homologous recombination including the RecBCD pathway.[66]

In this pathway, a three-subunit enzyme complex called RecBCD initiates recombination by binding to a blunt or nearly blunt end of a break in double-strand DNA. After RecBCD binds the DNA end, the RecB and RecD subunits begin unzipping the DNA duplex through helicase activity. The RecB subunit also has a nuclease domain, which cuts the single strand of DNA that emerges from the unzipping process. This unzipping continues until RecBCD encounters a specific nucleotide sequence (5'-GCTGGTGG-3') known as a Chi site.[65]

Upon encountering a Chi site, the activity of the RecBCD enzyme changes drastically.[64][61][67] DNA unwinding pauses for a few seconds and then resumes at roughly half the initial speed. This is likely because the slower RecB helicase unwinds the DNA after Chi, rather than the faster RecD helicase, which unwinds the DNA before Chi.[68][69] Recognition of the Chi site also changes the RecBCD enzyme so that it cuts the DNA strand with Chi and begins loading multiple RecA proteins onto the single-stranded DNA with the newly generated 3' end. The resulting RecA-coated nucleoprotein filament then searches out similar sequences of DNA on a homologous chromosome. The search process induces stretching of the DNA duplex, which enhances homology recognition (a mechanism termed conformational proofreading[70][71][72]). Upon finding such a sequence, the single-stranded nucleoprotein filament moves into the homologous recipient DNA duplex in a process called strand invasion.[73] The invading 3' overhang causes one of the strands of the recipient DNA duplex to be displaced, to form a D-loop. If the D-loop is cut, another swapping of strands forms a cross-shaped structure called a Holliday junction.[65] Resolution of the Holliday junction by some combination of RuvABC or RecG can produce two recombinant DNA molecules with reciprocal genetic types, if the two interacting DNA molecules differ genetically. Alternatively, the invading 3’ end near Chi can prime DNA synthesis and form a replication fork. This type of resolution produces only one type of recombinant (non-reciprocal).

RecF pathway edit

Bacteria appear to use the RecF pathway of homologous recombination to repair single-strand gaps in DNA. When the RecBCD pathway is inactivated by mutations and additional mutations inactivate the SbcCD and ExoI nucleases, the RecF pathway can also repair DNA double-strand breaks.[74] In the RecF pathway the RecQ helicase unwinds the DNA and the RecJ nuclease degrades the strand with a 5' end, leaving the strand with the 3' end intact. RecA protein binds to this strand and is either aided by the RecF, RecO, and RecR proteins or stabilized by them. The RecA nucleoprotein filament then searches for a homologous DNA and exchanges places with the identical or nearly identical strand in the homologous DNA.

Although the proteins and specific mechanisms involved in their initial phases differ, the two pathways are similar in that they both require single-stranded DNA with a 3' end and the RecA protein for strand invasion. The pathways are also similar in their phases of branch migration, in which the Holliday junction slides in one direction, and resolution, in which the Holliday junctions are cleaved apart by enzymes.[75][76] The alternative, non-reciprocal type of resolution may also occur by either pathway.

Branch migration edit

Immediately after strand invasion, the Holliday junction moves along the linked DNA during the branch migration process. It is in this movement of the Holliday junction that base pairs between the two homologous DNA duplexes are exchanged. To catalyze branch migration, the RuvA protein first recognizes and binds to the Holliday junction and recruits the RuvB protein to form the RuvAB complex. Two sets of the RuvB protein, which each form a ring-shaped ATPase, are loaded onto opposite sides of the Holliday junction, where they act as twin pumps that provide the force for branch migration. Between those two rings of RuvB, two sets of the RuvA protein assemble in the center of the Holliday junction such that the DNA at the junction is sandwiched between each set of RuvA. The strands of both DNA duplexes—the "donor" and the "recipient" duplexes—are unwound on the surface of RuvA as they are guided by the protein from one duplex to the other.[77][78]

Resolution edit

In the resolution phase of recombination, any Holliday junctions formed by the strand invasion process are cut, thereby restoring two separate DNA molecules. This cleavage is done by RuvAB complex interacting with RuvC, which together form the RuvABC complex. RuvC is an endonuclease that cuts the degenerate sequence 5'-(A/T)TT(G/C)-3'. The sequence is found frequently in DNA, about once every 64 nucleotides.[78] Before cutting, RuvC likely gains access to the Holliday junction by displacing one of the two RuvA tetramers covering the DNA there.[77] Recombination results in either "splice" or "patch" products, depending on how RuvC cleaves the Holliday junction.[78] Splice products are crossover products, in which there is a rearrangement of genetic material around the site of recombination. Patch products, on the other hand, are non-crossover products in which there is no such rearrangement and there is only a "patch" of hybrid DNA in the recombination product.[79]

Facilitating genetic transfer edit

Homologous recombination is an important method of integrating donor DNA into a recipient organism's genome in horizontal gene transfer, the process by which an organism incorporates foreign DNA from another organism without being the offspring of that organism. Homologous recombination requires incoming DNA to be highly similar to the recipient genome, and so horizontal gene transfer is usually limited to similar bacteria.[80] Studies in several species of bacteria have established that there is a log-linear decrease in recombination frequency with increasing difference in sequence between host and recipient DNA.[81][82][83]

In bacterial conjugation, where DNA is transferred between bacteria through direct cell-to-cell contact, homologous recombination helps integrate foreign DNA into the host genome via the RecBCD pathway. The RecBCD enzyme promotes recombination after DNA is converted from single-strand DNA–in which form it originally enters the bacterium–to double-strand DNA during replication. The RecBCD pathway is also essential for the final phase of transduction, a type of horizontal gene transfer in which DNA is transferred from one bacterium to another by a virus. Foreign, bacterial DNA is sometimes misincorporated in the capsid head of bacteriophage virus particles as DNA is packaged into new bacteriophages during viral replication. When these new bacteriophages infect other bacteria, DNA from the previous host bacterium is injected into the new bacterial host as double-strand DNA. The RecBCD enzyme then incorporates this double-strand DNA into the genome of the new bacterial host.[65]

Bacterial transformation edit

Natural bacterial transformation involves the transfer of DNA from a donor bacterium to a recipient bacterium, where both donor and recipient are ordinarily of the same species. Transformation, unlike bacterial conjugation and transduction, depends on numerous bacterial gene products that specifically interact to perform this process.[84] Thus transformation is clearly a bacterial adaptation for DNA transfer. In order for a bacterium to bind, take up and integrate donor DNA into its resident chromosome by homologous recombination, it must first enter a special physiological state termed competence. The RecA/Rad51/DMC1 gene family plays a central role in homologous recombination during bacterial transformation as it does during eukaryotic meiosis and mitosis. For instance, the RecA protein is essential for transformation in Bacillus subtilis and Streptococcus pneumoniae,[85] and expression of the RecA gene is induced during the development of competence for transformation in these organisms.

As part of the transformation process, the RecA protein interacts with entering single-stranded DNA (ssDNA) to form RecA/ssDNA nucleofilaments that scan the resident chromosome for regions of homology and bring the entering ssDNA to the corresponding region, where strand exchange and homologous recombination occur.[86] Thus the process of homologous recombination during bacterial transformation has fundamental similarities to homologous recombination during meiosis.

In viruses edit

Homologous recombination occurs in several groups of viruses. In DNA viruses such as herpesvirus, recombination occurs through a break-and-rejoin mechanism like in bacteria and eukaryotes.[87] There is also evidence for recombination in some RNA viruses, specifically positive-sense ssRNA viruses like retroviruses, picornaviruses, and coronaviruses. There is controversy over whether homologous recombination occurs in negative-sense ssRNA viruses like influenza.[88]

In RNA viruses, homologous recombination can be either precise or imprecise. In the precise type of RNA-RNA recombination, there is no difference between the two parental RNA sequences and the resulting crossover RNA region. Because of this, it is often difficult to determine the location of crossover events between two recombining RNA sequences. In imprecise RNA homologous recombination, the crossover region has some difference with the parental RNA sequences – caused by either addition, deletion, or other modification of nucleotides. The level of precision in crossover is controlled by the sequence context of the two recombining strands of RNA: sequences rich in adenine and uracil decrease crossover precision.[89][90]

Homologous recombination is important in facilitating viral evolution.[89][91] For example, if the genomes of two viruses with different disadvantageous mutations undergo recombination, then they may be able to regenerate a fully functional genome. Alternatively, if two similar viruses have infected the same host cell, homologous recombination can allow those two viruses to swap genes and thereby evolve more potent variations of themselves.[91]

Homologous recombination is the proposed mechanism whereby the DNA virus human herpesvirus-6 integrates into human telomeres.[92]

When two or more viruses, each containing lethal genomic damage, infect the same host cell, the virus genomes can often pair with each other and undergo homologous recombinational repair to produce viable progeny. This process, known as multiplicity reactivation, has been studied in several bacteriophages, including phage T4.[93] Enzymes employed in recombinational repair in phage T4 are functionally homologous to enzymes employed in bacterial and eukaryotic recombinational repair.[94] In particular, with regard to a gene necessary for the strand exchange reaction, a key step in homologous recombinational repair, there is functional homology from viruses to humans (i. e. uvsX in phage T4; recA in E. coli and other bacteria, and rad51 and dmc1 in yeast and other eukaryotes, including humans).[95] Multiplicity reactivation has also been demonstrated in numerous pathogenic viruses.[96]

Coronavirus edit

Coronaviruses are capable of genetic recombination when at least two viral genomes are present in the same infected cell. RNA recombination appears to be a major driving force in determining (1) genetic variability within a CoV species, (2) the capability of a CoV species to jump from one host to another, and (3) infrequently, the emergence of novel CoVs.[97] The mechanism of recombination in CoVs likely involves template switching during genome replication.[97] Recombination in RNA viruses appears to be an adaptation for coping with genome damage.[98]

The pandemic SARS-CoV-2's entire receptor binding motif appears to have been introduced through recombination from coronaviruses of pangolins.[99] Such a recombination event may have been a critical step in the evolution of SARS-CoV-2's capability to infect humans.[99] Recombination events are likely key steps in the evolutionary process that leads to the emergence of new human coronaviruses.[100]

During COVID-19 pandemic in 2020, many genomic sequences of Australian SARS‐CoV‐2 isolates have deletions or mutations (29742G>A or 29742G>U; "G19A" or "G19U")in the Coronavirus 3′ stem-loop II-like motif (s2m), an RNA motif in 3' untranslated region of viral genome, suggesting that RNA recombination events may have occurred in s2m of SARS-CoV-2. Based on computational analysis of 1319 Australia SARS‐CoV‐2 sequences using Recco algorithm (https://recco.bioinf.mpi-inf.mpg.de/), 29742G("G19"), 29744G("G21"), and 29751G("G28") were predicted as recombination hotspots.[101]

 
Schematic representation of the s2m RNA secondary structure, with tertiary structural interactions indicated as long range contacts.

The SARS-CoV-2 outbreak in Diamond Princess cruise most likely originated from either a single person infected with a virus variant identical to the Wuhan WIV04 isolates, or simultaneously with another primary case infected with a virus containing the 11083G > T mutation. Linkage disequilibrium analysis confirmed that RNA recombination with the 11083G > T mutation also contributed to the increase of mutations among the viral progeny. The findings indicate that the 11083G > T mutation of SARS-CoV-2 spread during shipboard quarantine and arose through de novo RNA recombination under positive selection pressure. In addition, in three patients in this cruise, two mutations 29736G > T and 29751G > T ("G13" and "G28") were also located in Coronavirus 3′ stem-loop II-like motif (s2m), as "G28" was predicted as recombination hotspots in Australian SARS-CoV-2 mutants. Although s2m is considered an RNA motif highly conserved among many coronavirus species, this result also suggests that s2m of SARS-CoV-2 is rather a RNA recombination/mutation hotspot.[102]

Effects of dysfunction edit

 
Figure 9. Joining of single-ended double strand breaks could lead to rearrangements

Without proper homologous recombination, chromosomes often incorrectly align for the first phase of cell division in meiosis. This causes chromosomes to fail to properly segregate in a process called nondisjunction. In turn, nondisjunction can cause sperm and ova to have too few or too many chromosomes. Down's syndrome, which is caused by an extra copy of chromosome 21, is one of many abnormalities that result from such a failure of homologous recombination in meiosis.[78][103]

Deficiencies in homologous recombination have been strongly linked to cancer formation in humans. For example, each of the cancer-related diseases Bloom syndrome, Werner syndrome and Rothmund–Thomson syndrome are caused by malfunctioning copies of RecQ helicase genes involved in the regulation of homologous recombination: BLM, WRN and RECQL4, respectively.[104] In the cells of Bloom's syndrome patients, who lack a working copy of the BLM protein, there is an elevated rate of homologous recombination.[105] Experiments in mice deficient in BLM have suggested that the mutation gives rise to cancer through a loss of heterozygosity caused by increased homologous recombination.[106] A loss in heterozygosity refers to the loss of one of two versions—or alleles—of a gene. If one of the lost alleles helps to suppress tumors, like the gene for the retinoblastoma protein for example, then the loss of heterozygosity can lead to cancer.[107]: 1236 

Decreased rates of homologous recombination cause inefficient DNA repair,[107]: 310  which can also lead to cancer.[108] This is the case with BRCA1 and BRCA2, two similar tumor suppressor genes whose malfunctioning has been linked with considerably increased risk for breast and ovarian cancer. Cells missing BRCA1 and BRCA2 have a decreased rate of homologous recombination and increased sensitivity to ionizing radiation, suggesting that decreased homologous recombination leads to increased susceptibility to cancer.[108] Because the only known function of BRCA2 is to help initiate homologous recombination, researchers have speculated that more detailed knowledge of BRCA2's role in homologous recombination may be the key to understanding the causes of breast and ovarian cancer.[108]

Tumours with a homologous recombination deficiency (including BRCA defects) are described as HRD-positive.[109]

Evolutionary conservation edit

 
Figure 10. Protein domains in homologous recombination-related proteins are conserved across the three main groups of life: archaea, bacteria and eukaryotes.

While the pathways can mechanistically vary, the ability of organisms to perform homologous recombination is universally conserved across all domains of life.[110] Based on the similarity of their amino acid sequences, homologs of a number of proteins can be found in multiple domains of life indicating that they evolved a long time ago, and have since diverged from common ancestral proteins.[110]

RecA recombinase family members are found in almost all organisms with RecA in bacteria, Rad51 and DMC1 in eukaryotes, RadA in archaea, and UvsX in T4 phage.[111]

Related single stranded binding proteins that are important for homologous recombination, and many other processes, are also found in all domains of life.[112]

Rad54, Mre11, Rad50, and a number of other proteins are also found in both archaea and eukaryotes.[110][111][113]

The RecA recombinase family edit

The proteins of the RecA recombinase family of proteins are thought to be descended from a common ancestral recombinase.[110] The RecA recombinase family contains RecA protein from bacteria, the Rad51 and Dmc1 proteins from eukaryotes, and RadA from archaea, and the recombinase paralog proteins. Studies modeling the evolutionary relationships between the Rad51, Dmc1 and RadA proteins indicate that they are monophyletic, or that they share a common molecular ancestor.[110] Within this protein family, Rad51 and Dmc1 are grouped together in a separate clade from RadA. One of the reasons for grouping these three proteins together is that they all possess a modified helix-turn-helix motif, which helps the proteins bind to DNA, toward their N-terminal ends.[110] An ancient gene duplication event of a eukaryotic RecA gene and subsequent mutation has been proposed as a likely origin of the modern RAD51 and DMC1 genes.[110]

The proteins generally share a long conserved region known as the RecA/Rad51 domain. Within this protein domain are two sequence motifs, Walker A motif and Walker B motif. The Walker A and B motifs allow members of the RecA/Rad51 protein family to engage in ATP binding and ATP hydrolysis.[110][114]

Meiosis-specific proteins edit

The discovery of Dmc1 in several species of Giardia, one of the earliest protists to diverge as a eukaryote, suggests that meiotic homologous recombination—and thus meiosis itself—emerged very early in eukaryotic evolution.[115] In addition to research on Dmc1, studies on the Spo11 protein have provided information on the origins of meiotic recombination.[116] Spo11, a type II topoisomerase, can initiate homologous recombination in meiosis by making targeted double-strand breaks in DNA.[25] Phylogenetic trees based on the sequence of genes similar to SPO11 in animals, fungi, plants, protists and archaea have led scientists to believe that the version Spo11 currently in eukaryotes emerged in the last common ancestor of eukaryotes and archaea.[116]

Technological applications edit

Gene targeting edit

 
Figure 11. As a developing embryo, this chimeric mouse had the agouti coat color gene introduced into its DNA via gene targeting. Its offspring are homozygous for the agouti gene.

Many methods for introducing DNA sequences into organisms to create recombinant DNA and genetically modified organisms use the process of homologous recombination.[117] Also called gene targeting, the method is especially common in yeast and mouse genetics. The gene targeting method in knockout mice uses mouse embryonic stem cells to deliver artificial genetic material (mostly of therapeutic interest), which represses the target gene of the mouse by the principle of homologous recombination. The mouse thereby acts as a working model to understand the effects of a specific mammalian gene. In recognition of their discovery of how homologous recombination can be used to introduce genetic modifications in mice through embryonic stem cells, Mario Capecchi, Martin Evans and Oliver Smithies were awarded the 2007 Nobel Prize for Physiology or Medicine.[118]

Advances in gene targeting technologies which hijack the homologous recombination mechanics of cells are now leading to the development of a new wave of more accurate, isogenic human disease models. These engineered human cell models are thought to more accurately reflect the genetics of human diseases than their mouse model predecessors. This is largely because mutations of interest are introduced into endogenous genes, just as they occur in the real patients, and because they are based on human genomes rather than rat genomes. Furthermore, certain technologies enable the knock-in of a particular mutation rather than just knock-outs associated with older gene targeting technologies.

Protein engineering edit

Protein engineering with homologous recombination develops chimeric proteins by swapping fragments between two parental proteins. These techniques exploit the fact that recombination can introduce a high degree of sequence diversity while preserving a protein's ability to fold into its tertiary structure, or three-dimensional shape.[119] This stands in contrast to other protein engineering techniques, like random point mutagenesis, in which the probability of maintaining protein function declines exponentially with increasing amino acid substitutions.[120] The chimeras produced by recombination techniques are able to maintain their ability to fold because their swapped parental fragments are structurally and evolutionarily conserved. These recombinable "building blocks" preserve structurally important interactions like points of physical contact between different amino acids in the protein's structure. Computational methods like SCHEMA and statistical coupling analysis can be used to identify structural subunits suitable for recombination.[121][122][123]

Techniques that rely on homologous recombination have been used to engineer new proteins.[121] In a study published in 2007, researchers were able to create chimeras of two enzymes involved in the biosynthesis of isoprenoids, a diverse class of compounds including hormones, visual pigments and certain pheromones. The chimeric proteins acquired an ability to catalyze an essential reaction in isoprenoid biosynthesis—one of the most diverse pathways of biosynthesis found in nature—that was absent in the parent proteins.[124] Protein engineering through recombination has also produced chimeric enzymes with new function in members of a group of proteins known as the cytochrome P450 family,[125] which in humans is involved in detoxifying foreign compounds like drugs, food additives and preservatives.[22]

Cancer therapy edit

Homologous recombination proficient (HRP) cancer cells are able to repair the DNA damage, which is caused by chemotherapy such as cisplatin. Thus, HRP cancers are difficult to treat. Studies suggest that homologous recombination can be targeted via c-Abl inhibition.[126][127] Cancer cells with BRCA mutations have deficiencies in homologous recombination, and drugs to exploit those deficiencies have been developed and used successfully in clinical trials.[128][129] Olaparib, a PARP1 inhibitor, shrunk or stopped the growth of tumors from breast, ovarian and prostate cancers caused by mutations in the BRCA1 or BRCA2 genes, which are necessary for HR. When BRCA1 or BRCA2 is absent, other types of DNA repair mechanisms must compensate for the deficiency of HR, such as base-excision repair (BER) for stalled replication forks or non-homologous end joining (NHEJ) for double strand breaks.[128] By inhibiting BER in an HR-deficient cell, olaparib applies the concept of synthetic lethality to specifically target cancer cells. While PARP1 inhibitors represent a novel approach to cancer therapy, researchers have cautioned that they may prove insufficient for treating late-stage metastatic cancers.[128] Cancer cells can become resistant to a PARP1 inhibitor if they undergo deletions of mutations in BRCA2, undermining the drug's synthetic lethality by restoring cancer cells' ability to repair DNA by HR.[130]

See also edit

References edit

  1. ^ Thompson LH, Schild D (June 2001). "Homologous recombinational repair of DNA ensures mammalian chromosome stability". Mutation Research. 477 (1–2): 131–53. doi:10.1016/S0027-5107(01)00115-4. PMID 11376695.
  2. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P, et al. (2002). "Chapter 5: DNA Replication, Repair, and Recombination". Molecular Biology of the Cell (4th ed.). New York: Garland Science. p. 845. ISBN 978-0-8153-3218-3. OCLC 145080076.
  3. ^ Capecchi MR (June 1989). "Altering the genome by homologous recombination". Science. 244 (4910): 1288–92. Bibcode:1989Sci...244.1288C. doi:10.1126/science.2660260. PMID 2660260.
  4. ^ Smithies O, Gregg RG, Boggs SS, Koralewski MA, Kucherlapati RS (1985-09-19). "Insertion of DNA sequences into the human chromosomal beta-globin locus by homologous recombination". Nature. 317 (6034): 230–4. Bibcode:1985Natur.317..230S. doi:10.1038/317230a0. PMID 2995814. S2CID 30212766.
  5. ^ Orr-Weaver TL, Szostak JW, Rothstein RJ (October 1981). "Yeast transformation: a model system for the study of recombination". Proceedings of the National Academy of Sciences of the United States of America. 78 (10): 6354–8. Bibcode:1981PNAS...78.6354O. doi:10.1073/pnas.78.10.6354. PMC 349037. PMID 6273866.
  6. ^ Orr-Weaver TL, Szostak JW (July 1983). "Yeast recombination: the association between double-strand gap repair and crossing-over". Proceedings of the National Academy of Sciences of the United States of America. 80 (14): 4417–21. Bibcode:1983PNAS...80.4417O. doi:10.1073/pnas.80.14.4417. PMC 384049. PMID 6308623.
  7. ^ a b Szostak JW, Orr-Weaver TL, Rothstein RJ, Stahl FW (May 1983). "The double-strand-break repair model for recombination". Cell. 33 (1): 25–35. doi:10.1016/0092-8674(83)90331-8. PMID 6380756. S2CID 39590123.
  8. ^ Resnick MA (June 1976). "The repair of double-strand breaks in DNA; a model involving recombination". Journal of Theoretical Biology. 59 (1): 97–106. Bibcode:1976JThBi..59...97R. doi:10.1016/s0022-5193(76)80025-2. PMID 940351.
  9. ^ Jasin M, Rothstein R (November 2013). "Repair of strand breaks by homologous recombination". Cold Spring Harbor Perspectives in Biology. 5 (11): a012740. doi:10.1101/cshperspect.a012740. PMC 3809576. PMID 24097900.
  10. ^ Bateson P (August 2002). "William Bateson: a biologist ahead of his time" (PDF). Journal of Genetics. 81 (2): 49–58. doi:10.1007/BF02715900. PMID 12532036. S2CID 26806110.
  11. ^ "Reginald Crundall Punnett". NAHSTE, University of Edinburgh. Retrieved 3 July 2010.
  12. ^ Lobo I, Shaw K (2008). "Thomas Hunt Morgan, genetic recombination, and gene mapping". Nature Education. 1 (1).
  13. ^ a b Coe E, Kass LB (May 2005). "Proof of physical exchange of genes on the chromosomes". Proceedings of the National Academy of Sciences of the United States of America. 102 (19): 6641–6. Bibcode:2005PNAS..102.6641C. doi:10.1073/pnas.0407340102. PMC 1100733. PMID 15867161.
  14. ^ Creighton HB, McClintock B (August 1931). "A Correlation of Cytological and Genetical Crossing-Over in Zea Mays". Proceedings of the National Academy of Sciences of the United States of America. 17 (8): 492–7. Bibcode:1931PNAS...17..492C. doi:10.1073/pnas.17.8.492. PMC 1076098. PMID 16587654.
  15. ^ Stern, C (1931). "Zytologisch-genetische untersuchungen alsbeweise fur die Morgansche theorie des faktoraustauschs". Biologisches Zentralblatt. 51: 547–587.
  16. ^ "The development of bacterial genetics". US National Library of Medicine. Retrieved 3 July 2010.
  17. ^ "The Nobel Prize in Physiology or Medicine 1958". Nobelprize.org. Retrieved 3 July 2010.
  18. ^ a b c Haber JE, Ira G, Malkova A, Sugawara N (January 2004). "Repairing a double-strand chromosome break by homologous recombination: revisiting Robin Holliday's model". Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 359 (1441): 79–86. doi:10.1098/rstb.2003.1367. PMC 1693306. PMID 15065659.
  19. ^ Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J (2000). "12.5: Recombination between Homologous DNA Sites: Double-Strand Breaks in DNA Initiate Recombination". Molecular Cell Biology (4th ed.). W. H. Freeman and Company. ISBN 978-0-7167-3136-8.
  20. ^ Griffiths A, et al. (1999). "8: Chromosome Mutations: Chromosomal Rearrangements". Modern Genetic Analysis. W. H. Freeman and Company. ISBN 978-0-7167-3118-4.
  21. ^ Khanna KK, Jackson SP (March 2001). "DNA double-strand breaks: signaling, repair and the cancer connection". Nature Genetics. 27 (3): 247–54. doi:10.1038/85798. PMID 11242102. S2CID 3012823.
  22. ^ a b c Nelson DL, Cox MM (2005). Principles of Biochemistry (4th ed.). Freeman. pp. 980–981. ISBN 978-0-7167-4339-2.
  23. ^ Marcon E, Moens PB (August 2005). "The evolution of meiosis: recruitment and modification of somatic DNA-repair proteins". BioEssays. 27 (8): 795–808. doi:10.1002/bies.20264. PMID 16015600. S2CID 27658497.
  24. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2008). Molecular Biology of the Cell (5th ed.). Garland Science. p. 305. ISBN 978-0-8153-4105-5.
  25. ^ a b Keeney S, Giroux CN, Kleckner N (February 1997). "Meiosis-specific DNA double-strand breaks are catalyzed by Spo11, a member of a widely conserved protein family". Cell. 88 (3): 375–84. doi:10.1016/S0092-8674(00)81876-0. PMID 9039264. S2CID 8294596.
  26. ^ Longhese MP, Bonetti D, Guerini I, Manfrini N, Clerici M (September 2009). "DNA double-strand breaks in meiosis: checking their formation, processing and repair". DNA Repair. 8 (9): 1127–38. doi:10.1016/j.dnarep.2009.04.005. PMID 19464965.
  27. ^ Cahill LP, Mariana JC, Mauléon P (January 1979). "Total follicular populations in ewes of high and low ovulation rates". Journal of Reproduction and Fertility. 55 (1): 27–36. doi:10.1530/jrf.0.0550027. PMID 423159.
  28. ^ Schimmel J, van Schendel R, den Dunnen JT, Tijsterman M (September 2019). "Templated Insertions: A Smoking Gun for Polymerase Theta-Mediated End Joining". Trends in Genetics. 35 (9): 632–644. doi:10.1016/j.tig.2019.06.001. PMID 31296341. S2CID 195892718.
  29. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2008). Molecular Biology of the Cell (5th ed.). Garland Science. p. 303. ISBN 978-0-8153-4105-5.
  30. ^ a b Shrivastav M, De Haro LP, Nickoloff JA (January 2008). "Regulation of DNA double-strand break repair pathway choice". Cell Research. 18 (1): 134–47. doi:10.1038/cr.2007.111. PMID 18157161.
  31. ^ a b Mimitou EP, Symington LS (May 2009). "Nucleases and helicases take center stage in homologous recombination". Trends in Biochemical Sciences. 34 (5): 264–72. doi:10.1016/j.tibs.2009.01.010. PMID 19375328.
  32. ^ Andres, Sara N.; Williams, R. Scott (August 2017). "CtIP/Ctp1/Sae2, molecular form fit for function". DNA Repair. 56: 109–117. doi:10.1016/j.dnarep.2017.06.013. PMC 5543718. PMID 28623092.
  33. ^ Huertas P, Cortés-Ledesma F, Sartori AA, Aguilera A, Jackson SP (October 2008). "CDK targets Sae2 to control DNA-end resection and homologous recombination". Nature. 455 (7213): 689–92. Bibcode:2008Natur.455..689H. doi:10.1038/nature07215. PMC 2635538. PMID 18716619.
  34. ^ Liu B, Yip RK, Zhou Z (2012). "Chromatin remodeling, DNA damage repair and aging". Curr. Genomics. 13 (7): 533–47. doi:10.2174/138920212803251373. PMC 3468886. PMID 23633913.
  35. ^ a b c d Sellou H, Lebeaupin T, Chapuis C, Smith R, Hegele A, Singh HR, Kozlowski M, Bultmann S, Ladurner AG, Timinszky G, Huet S (2016). "The poly(ADP-ribose)-dependent chromatin remodeler Alc1 induces local chromatin relaxation upon DNA damage". Mol. Biol. Cell. 27 (24): 3791–3799. doi:10.1091/mbc.E16-05-0269. PMC 5170603. PMID 27733626.
  36. ^ a b Van Meter M, Simon M, Tombline G, May A, Morello TD, Hubbard BP, Bredbenner K, Park R, Sinclair DA, Bohr VA, Gorbunova V, Seluanov A (2016). "JNK Phosphorylates SIRT6 to Stimulate DNA Double-Strand Break Repair in Response to Oxidative Stress by Recruiting PARP1 to DNA Breaks". Cell Rep. 16 (10): 2641–50. doi:10.1016/j.celrep.2016.08.006. PMC 5089070. PMID 27568560.
  37. ^ a b Haince JF, McDonald D, Rodrigue A, Déry U, Masson JY, Hendzel MJ, Poirier GG (2008). "PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites". J. Biol. Chem. 283 (2): 1197–208. doi:10.1074/jbc.M706734200. PMID 18025084.
  38. ^ a b c Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM (1998). "DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139". J. Biol. Chem. 273 (10): 5858–68. doi:10.1074/jbc.273.10.5858. PMID 9488723.
  39. ^ Mailand N, Bekker-Jensen S, Faustrup H, Melander F, Bartek J, Lukas C, Lukas J (2007). "RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins". Cell. 131 (5): 887–900. doi:10.1016/j.cell.2007.09.040. PMID 18001824. S2CID 14232192.
  40. ^ Luijsterburg MS, Acs K, Ackermann L, Wiegant WW, Bekker-Jensen S, Larsen DH, Khanna KK, van Attikum H, Mailand N, Dantuma NP (2012). "A new non-catalytic role for ubiquitin ligase RNF8 in unfolding higher-order chromatin structure". EMBO J. 31 (11): 2511–27. doi:10.1038/emboj.2012.104. PMC 3365417. PMID 22531782.
  41. ^ Baudat F, Buard J, Grey C, Fledel-Alon A, Ober C, Przeworski M, et al. (February 2010). "PRDM9 is a major determinant of meiotic recombination hotspots in humans and mice". Science. 327 (5967): 836–40. Bibcode:2010Sci...327..836B. doi:10.1126/science.1183439. PMC 4295902. PMID 20044539.
  42. ^ Wells D, Bitoun E, Moralli D, Zhang G, Hinch A, Jankowska J, et al. (August 2020). "ZCWPW1 is recruited to recombination hotspots by PRDM9, and is essential for meiotic double strand break repair". eLife. 9: e53392. doi:10.7554/eLife.53392. PMC 7494361. PMID 32744506.
  43. ^ a b c d Sung P, Klein H (October 2006). "Mechanism of homologous recombination: mediators and helicases take on regulatory functions". Nature Reviews Molecular Cell Biology. 7 (10): 739–50. doi:10.1038/nrm2008. PMID 16926856. S2CID 30324005.
  44. ^ Wold MS (1997). "Replication protein A: a heterotrimeric, single-stranded DNA-binding protein required for eukaryotic DNA metabolism". Annual Review of Biochemistry. 66: 61–92. doi:10.1146/annurev.biochem.66.1.61. PMID 9242902.
  45. ^ McMahill MS, Sham CW, Bishop DK (November 2007). "Synthesis-dependent strand annealing in meiosis". PLOS Biology. 5 (11): e299. doi:10.1371/journal.pbio.0050299. PMC 2062477. PMID 17988174.  
  46. ^ Bärtsch S, Kang LE, Symington LS (February 2000). "RAD51 is required for the repair of plasmid double-stranded DNA gaps from either plasmid or chromosomal templates". Molecular and Cellular Biology. 20 (4): 1194–205. doi:10.1128/MCB.20.4.1194-1205.2000. PMC 85244. PMID 10648605.
  47. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2008). Molecular Biology of the Cell (5th ed.). Garland Science. pp. 312–313. ISBN 978-0-8153-4105-5.
  48. ^ a b c Helleday T, Lo J, van Gent DC, Engelward BP (July 2007). "DNA double-strand break repair: from mechanistic understanding to cancer treatment". DNA Repair. 6 (7): 923–35. doi:10.1016/j.dnarep.2007.02.006. PMID 17363343.
  49. ^ a b Andersen SL, Sekelsky J (December 2010). "Meiotic versus mitotic recombination: two different routes for double-strand break repair: the different functions of meiotic versus mitotic DSB repair are reflected in different pathway usage and different outcomes". BioEssays. 32 (12): 1058–66. doi:10.1002/bies.201000087. PMC 3090628. PMID 20967781.
  50. ^ Allers T, Lichten M (July 2001). "Differential timing and control of noncrossover and crossover recombination during meiosis". Cell. 106 (1): 47–57. doi:10.1016/s0092-8674(01)00416-0. PMID 11461701. S2CID 1878863.
  51. ^ Haber lab. "Single-strand annealing". Brandeis University. Retrieved 3 July 2010.
  52. ^ a b c Lyndaker AM, Alani E (March 2009). "A tale of tails: insights into the coordination of 3' end processing during homologous recombination". BioEssays. 31 (3): 315–21. doi:10.1002/bies.200800195. PMC 2958051. PMID 19260026.
  53. ^ Mimitou EP, Symington LS (September 2009). "DNA end resection: many nucleases make light work". DNA Repair. 8 (9): 983–95. doi:10.1016/j.dnarep.2009.04.017. PMC 2760233. PMID 19473888.
  54. ^ Pâques F, Haber JE (June 1999). "Multiple pathways of recombination induced by double-strand breaks in Saccharomyces cerevisiae". Microbiology and Molecular Biology Reviews. 63 (2): 349–404. doi:10.1128/MMBR.63.2.349-404.1999. PMC 98970. PMID 10357855.
  55. ^ a b McEachern MJ, Haber JE (2006). "Break-induced replication and recombinational telomere elongation in yeast". Annual Review of Biochemistry. 75: 111–35. doi:10.1146/annurev.biochem.74.082803.133234. PMID 16756487.
  56. ^ Morrish TA, Greider CW (January 2009). Haber JE (ed.). "Short telomeres initiate telomere recombination in primary and tumor cells". PLOS Genetics. 5 (1): e1000357. doi:10.1371/journal.pgen.1000357. PMC 2627939. PMID 19180191.  
  57. ^ Muntoni A, Reddel RR (October 2005). "The first molecular details of ALT in human tumor cells". Human Molecular Genetics. 14 Spec No. 2 (Review Issue 2): R191–6. doi:10.1093/hmg/ddi266. PMID 16244317.
  58. ^ PDB: 3cmt​; Chen Z, Yang H, Pavletich NP (May 2008). "Mechanism of homologous recombination from the RecA-ssDNA/dsDNA structures". Nature. 453 (7194): 489–4. Bibcode:2008Natur.453..489C. doi:10.1038/nature06971. PMID 18497818. S2CID 4416531.
  59. ^ Kowalczykowski SC, Dixon DA, Eggleston AK, Lauder SD, Rehrauer WM (September 1994). "Biochemistry of homologous recombination in Escherichia coli". Microbiological Reviews. 58 (3): 401–65. doi:10.1128/MMBR.58.3.401-465.1994. PMC 372975. PMID 7968921.
  60. ^ Rocha EP, Cornet E, Michel B (August 2005). "Comparative and evolutionary analysis of the bacterial homologous recombination systems". PLOS Genetics. 1 (2): e15. doi:10.1371/journal.pgen.0010015. PMC 1193525. PMID 16132081.  
  61. ^ a b Amundsen SK, Taylor AF, Reddy M, Smith GR (December 2007). "Intersubunit signaling in RecBCD enzyme, a complex protein machine regulated by Chi hot spots". Genes & Development. 21 (24): 3296–307. doi:10.1101/gad.1605807. PMC 2113030. PMID 18079176.
  62. ^ Singleton MR, Dillingham MS, Gaudier M, Kowalczykowski SC, Wigley DB (November 2004). (PDF). Nature. 432 (7014): 187–93. Bibcode:2004Natur.432..187S. doi:10.1038/nature02988. PMID 15538360. S2CID 2916995. Archived from the original (PDF) on 2004-05-25.
  63. ^ Cromie GA (August 2009). "Phylogenetic ubiquity and shuffling of the bacterial RecBCD and AddAB recombination complexes". Journal of Bacteriology. 191 (16): 5076–84. doi:10.1128/JB.00254-09. PMC 2725590. PMID 19542287.
  64. ^ a b Smith GR (June 2012). "How RecBCD enzyme and Chi promote DNA break repair and recombination: a molecular biologist's view". Microbiology and Molecular Biology Reviews. 76 (2): 217–28. doi:10.1128/MMBR.05026-11. PMC 3372252. PMID 22688812.
  65. ^ a b c d Dillingham MS, Kowalczykowski SC (December 2008). "RecBCD enzyme and the repair of double-stranded DNA breaks". Microbiology and Molecular Biology Reviews. 72 (4): 642–71, Table of Contents. doi:10.1128/MMBR.00020-08. PMC 2593567. PMID 19052323.
  66. ^ Michel B, Boubakri H, Baharoglu Z, LeMasson M, Lestini R (July 2007). "Recombination proteins and rescue of arrested replication forks". DNA Repair. 6 (7): 967–80. doi:10.1016/j.dnarep.2007.02.016. PMID 17395553.
  67. ^ Spies M, Bianco PR, Dillingham MS, Handa N, Baskin RJ, Kowalczykowski SC (September 2003). "A molecular throttle: the recombination hotspot chi controls DNA translocation by the RecBCD helicase". Cell. 114 (5): 647–54. doi:10.1016/S0092-8674(03)00681-0. PMID 13678587. S2CID 16662983.
  68. ^ Taylor AF, Smith GR (June 2003). "RecBCD enzyme is a DNA helicase with fast and slow motors of opposite polarity". Nature. 423 (6942): 889–93. Bibcode:2003Natur.423..889T. doi:10.1038/nature01674. PMID 12815437. S2CID 4302346.
  69. ^ Spies M, Amitani I, Baskin RJ, Kowalczykowski SC (November 2007). "RecBCD enzyme switches lead motor subunits in response to chi recognition". Cell. 131 (4): 694–705. doi:10.1016/j.cell.2007.09.023. PMC 2151923. PMID 18022364.
  70. ^ Savir Y, Tlusty T (November 2010). (PDF). Molecular Cell. 40 (3): 388–96. arXiv:1011.4382. Bibcode:2010arXiv1011.4382S. doi:10.1016/j.molcel.2010.10.020. PMID 21070965. S2CID 1682936. Archived from the original (PDF) on 2012-10-07. Retrieved 2011-08-31.
  71. ^ Rambo RP, Williams GJ, Tainer JA (November 2010). (PDF). Molecular Cell. 40 (3): 347–8. doi:10.1016/j.molcel.2010.10.032. PMC 3003302. PMID 21070960. Archived from the original (PDF) on 2012-10-07. Retrieved 2011-08-31.
  72. ^ De Vlaminck I, van Loenhout MT, Zweifel L, den Blanken J, Hooning K, Hage S, et al. (June 2012). "Mechanism of homology recognition in DNA recombination from dual-molecule experiments". Molecular Cell. 46 (5): 616–24. doi:10.1016/j.molcel.2012.03.029. PMID 22560720.
  73. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2008). Molecular Biology of the Cell (5th ed.). Garland Science. p. 307. ISBN 978-0-8153-4105-5.
  74. ^ Morimatsu K, Kowalczykowski SC (May 2003). "RecFOR proteins load RecA protein onto gapped DNA to accelerate DNA strand exchange: a universal step of recombinational repair". Molecular Cell. 11 (5): 1337–47. doi:10.1016/S1097-2765(03)00188-6. PMID 12769856.
  75. ^ Hiom K (July 2009). "DNA repair: common approaches to fixing double-strand breaks". Current Biology. 19 (13): R523–5. doi:10.1016/j.cub.2009.06.009. PMID 19602417. S2CID 2221866.
  76. ^ Handa N, Morimatsu K, Lovett ST, Kowalczykowski SC (May 2009). "Reconstitution of initial steps of dsDNA break repair by the RecF pathway of E. coli". Genes & Development. 23 (10): 1234–45. doi:10.1101/gad.1780709. PMC 2685532. PMID 19451222.
  77. ^ a b West SC (June 2003). "Molecular views of recombination proteins and their control". Nature Reviews Molecular Cell Biology. 4 (6): 435–45. doi:10.1038/nrm1127. PMID 12778123. S2CID 28474965.
  78. ^ a b c d Watson JD, Baker TA, Bell SP, Gann A, Levine M, Losick R (2003). Molecular Biology of the Gene (5th ed.). Pearson/Benjamin Cummings. pp. 259–291. ISBN 978-0-8053-4635-0.
  79. ^ Gumbiner-Russo LM, Rosenberg SM (28 November 2007). Sandler S (ed.). "Physical analyses of E. coli heteroduplex recombination products in vivo: on the prevalence of 5' and 3' patches". PLOS ONE. 2 (11): e1242. Bibcode:2007PLoSO...2.1242G. doi:10.1371/journal.pone.0001242. PMC 2082072. PMID 18043749.  
  80. ^ Thomas CM, Nielsen KM (September 2005). (PDF). Nature Reviews. Microbiology. 3 (9): 711–21. doi:10.1038/nrmicro1234. PMID 16138099. S2CID 1231127. Archived from the original (PDF) on 2010-06-01.
  81. ^ Vulić M, Dionisio F, Taddei F, Radman M (September 1997). "Molecular keys to speciation: DNA polymorphism and the control of genetic exchange in enterobacteria". Proceedings of the National Academy of Sciences of the United States of America. 94 (18): 9763–7. Bibcode:1997PNAS...94.9763V. doi:10.1073/pnas.94.18.9763. PMC 23264. PMID 9275198.
  82. ^ Majewski J, Cohan FM (January 1998). "The effect of mismatch repair and heteroduplex formation on sexual isolation in Bacillus". Genetics. 148 (1): 13–8. doi:10.1093/genetics/148.1.13. PMC 1459767. PMID 9475717.
  83. ^ Majewski J, Zawadzki P, Pickerill P, Cohan FM, Dowson CG (February 2000). "Barriers to genetic exchange between bacterial species: Streptococcus pneumoniae transformation". Journal of Bacteriology. 182 (4): 1016–23. doi:10.1128/JB.182.4.1016-1023.2000. PMC 94378. PMID 10648528.
  84. ^ Chen I, Dubnau D (March 2004). "DNA uptake during bacterial transformation". Nature Reviews. Microbiology. 2 (3): 241–9. doi:10.1038/nrmicro844. PMID 15083159. S2CID 205499369.
  85. ^ Claverys JP, Martin B, Polard P (May 2009). "The genetic transformation machinery: composition, localization, and mechanism". FEMS Microbiology Reviews. 33 (3): 643–56. doi:10.1111/j.1574-6976.2009.00164.x. PMID 19228200.
  86. ^ Kidane D, Graumann PL (July 2005). "Intracellular protein and DNA dynamics in competent Bacillus subtilis cells". Cell. 122 (1): 73–84. doi:10.1016/j.cell.2005.04.036. PMID 16009134. S2CID 17272331.
  87. ^ Fleischmann Jr WR (1996). "43". Medical Microbiology (4th ed.). University of Texas Medical Branch at Galveston. ISBN 978-0-9631172-1-2.
  88. ^ Boni MF, de Jong MD, van Doorn HR, Holmes EC (3 May 2010). Martin DP (ed.). "Guidelines for identifying homologous recombination events in influenza A virus". PLOS ONE. 5 (5): e10434. Bibcode:2010PLoSO...510434B. doi:10.1371/journal.pone.0010434. PMC 2862710. PMID 20454662.  
  89. ^ a b Nagy PD, Bujarski JJ (January 1996). "Homologous RNA recombination in brome mosaic virus: AU-rich sequences decrease the accuracy of crossovers". Journal of Virology. 70 (1): 415–26. doi:10.1128/JVI.70.1.415-426.1996. PMC 189831. PMID 8523555.
  90. ^ Chetverin AB (October 1999). "The puzzle of RNA recombination". FEBS Letters. 460 (1): 1–5. doi:10.1016/S0014-5793(99)01282-X. PMC 7163957. PMID 10571050.
  91. ^ a b Roossinck MJ (September 1997). "Mechanisms of plant virus evolution". Annual Review of Phytopathology. 35: 191–209. doi:10.1146/annurev.phyto.35.1.191. PMID 15012521.
  92. ^ Arbuckle JH, Medveczky PG (August 2011). "The molecular biology of human herpesvirus-6 latency and telomere integration". Microbes and Infection / Institut Pasteur. 13 (8–9): 731–41. doi:10.1016/j.micinf.2011.03.006. PMC 3130849. PMID 21458587.
  93. ^ Bernstein C (March 1981). "Deoxyribonucleic acid repair in bacteriophage". Microbiological Reviews. 45 (1): 72–98. doi:10.1128/MMBR.45.1.72-98.1981. PMC 281499. PMID 6261109.
  94. ^ Bernstein C, Bernstein H (2001). DNA repair in bacteriophage. In: Nickoloff JA, Hoekstra MF (Eds.) DNA Damage and Repair, Vol.3. Advances from Phage to Humans. Humana Press, Totowa, NJ, pp. 1–19. ISBN 978-0896038035
  95. ^ Story RM, Bishop DK, Kleckner N, Steitz TA (March 1993). "Structural relationship of bacterial RecA proteins to recombination proteins from bacteriophage T4 and yeast". Science. 259 (5103): 1892–6. Bibcode:1993Sci...259.1892S. doi:10.1126/science.8456313. PMID 8456313.
  96. ^ Michod RE, Bernstein H, Nedelcu AM (May 2008). "Adaptive value of sex in microbial pathogens". Infection, Genetics and Evolution. 8 (3): 267–85. doi:10.1016/j.meegid.2008.01.002. PMID 18295550.http://www.hummingbirds.arizona.edu/Faculty/Michod/Downloads/IGE%20review%20sex.pdf
  97. ^ a b Su, Shuo; Wong, Gary; Shi, Weifeng; Liu, Jun; Lai, Alexander C.K.; Zhou, Jiyong; Liu, Wenjun; Bi, Yuhai; Gao, George F. (2016). "Epidemiology, Genetic Recombination, and Pathogenesis of Coronaviruses". Trends in Microbiology. 24 (6): 490–502. doi:10.1016/j.tim.2016.03.003. PMC 7125511. PMID 27012512.
  98. ^ Barr, J. N.; Fearns, R. (2010). "How RNA viruses maintain their genome integrity". Journal of General Virology. 91 (6): 1373–1387. doi:10.1099/vir.0.020818-0. PMID 20335491.
  99. ^ a b Li, Xiaojun; Giorgi, Elena E.; Marichannegowda, Manukumar Honnayakanahalli; Foley, Brian; Xiao, Chuan; Kong, Xiang-Peng; Chen, Yue; Gnanakaran, S.; Korber, Bette; Gao, Feng (2020). "Emergence of SARS-CoV-2 through recombination and strong purifying selection". Science Advances. 6 (27). Bibcode:2020SciA....6.9153L. doi:10.1126/sciadv.abb9153. PMC 7458444. PMID 32937441.
  100. ^ Rehman, Saif ur; Shafique, Laiba; Ihsan, Awais; Liu, Qingyou (2020). "Evolutionary Trajectory for the Emergence of Novel Coronavirus SARS-CoV-2". Pathogens. 9 (3): 240. doi:10.3390/pathogens9030240. PMC 7157669. PMID 32210130.
  101. ^ Yeh TY, Contreras GP (July 2020). "Emerging viral mutants in Australia suggest RNA recombination event in the SARS-CoV-2 genome". The Medical Journal of Australia. 213 (1): 44–44.e1. doi:10.5694/mja2.50657. PMC 7300921. PMID 32506536.
  102. ^ Yeh TY, Contreras GP (1 July 2021). "Viral transmission and evolution dynamics of SARS-CoV-2 in shipboard quarantine". Bull. World Health Organ. 99 (7): 486–495. doi:10.2471/BLT.20.255752. PMC 8243027. PMID 34248221.
  103. ^ Lamb NE, Yu K, Shaffer J, Feingold E, Sherman SL (January 2005). "Association between maternal age and meiotic recombination for trisomy 21". American Journal of Human Genetics. 76 (1): 91–9. doi:10.1086/427266. PMC 1196437. PMID 15551222.
  104. ^ Cold Spring Harbor Laboratory (2007). "Human RecQ Helicases, Homologous Recombination And Genomic Instability". ScienceDaily. Retrieved 3 July 2010.
  105. ^ Modesti M, Kanaar R (2001). "Homologous recombination: from model organisms to human disease". Genome Biology. 2 (5): REVIEWS1014. doi:10.1186/gb-2001-2-5-reviews1014. PMC 138934. PMID 11387040.
  106. ^ Luo G, Santoro IM, McDaniel LD, Nishijima I, Mills M, Youssoufian H, Vogel H, Schultz RA, Bradley A (December 2000). "Cancer predisposition caused by elevated mitotic recombination in Bloom mice". Nature Genetics. 26 (4): 424–9. doi:10.1038/82548. PMID 11101838. S2CID 21218975.
  107. ^ a b Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2007). Molecular Biology of the Cell (5th ed.). Garland Science. ISBN 978-0-8153-4110-9.
  108. ^ a b c Powell SN, Kachnic LA (September 2003). "Roles of BRCA1 and BRCA2 in homologous recombination, DNA replication fidelity and the cellular response to ionizing radiation". Oncogene. 22 (37): 5784–91. doi:10.1038/sj.onc.1206678. PMID 12947386.
  109. ^ . Archived from the original on 2017-04-30. Retrieved 2016-12-30.
  110. ^ a b c d e f g h Lin Z, Kong H, Nei M, Ma H (July 2006). "Origins and evolution of the recA/RAD51 gene family: evidence for ancient gene duplication and endosymbiotic gene transfer". Proceedings of the National Academy of Sciences of the United States of America. 103 (27): 10328–33. Bibcode:2006PNAS..10310328L. doi:10.1073/pnas.0604232103. PMC 1502457. PMID 16798872.
  111. ^ a b Haseltine CA, Kowalczykowski SC (May 2009). "An archaeal Rad54 protein remodels DNA and stimulates DNA strand exchange by RadA". Nucleic Acids Research. 37 (8): 2757–70. doi:10.1093/nar/gkp068. PMC 2677860. PMID 19282450.
  112. ^ Rolfsmeier ML, Haseltine CA (March 2010). "The single-stranded DNA binding protein of Sulfolobus solfataricus acts in the presynaptic step of homologous recombination". Journal of Molecular Biology. 397 (1): 31–45. doi:10.1016/j.jmb.2010.01.004. PMID 20080104.
  113. ^ Huang Q, Liu L, Liu J, Ni J, She Q, Shen Y (2015). "Efficient 5'-3' DNA end resection by HerA and NurA is essential for cell viability in the crenarchaeon Sulfolobus islandicus". BMC Molecular Biology. 16: 2. doi:10.1186/s12867-015-0030-z. PMC 4351679. PMID 25880130.  
  114. ^ Jain SK, Cox MM, Inman RB (August 1994). "On the role of ATP hydrolysis in RecA protein-mediated DNA strand exchange. III. Unidirectional branch migration and extensive hybrid DNA formation". The Journal of Biological Chemistry. 269 (32): 20653–61. doi:10.1016/S0021-9258(17)32043-4. PMID 8051165.
  115. ^ Ramesh MA, Malik SB, Logsdon JM (January 2005). "A phylogenomic inventory of meiotic genes; evidence for sex in Giardia and an early eukaryotic origin of meiosis". Current Biology. 15 (2): 185–91. doi:10.1016/j.cub.2005.01.003. PMID 15668177. S2CID 17013247.
  116. ^ a b Malik SB, Ramesh MA, Hulstrand AM, Logsdon JM (December 2007). "Protist homologs of the meiotic Spo11 gene and topoisomerase VI reveal an evolutionary history of gene duplication and lineage-specific loss". Molecular Biology and Evolution. 24 (12): 2827–41. doi:10.1093/molbev/msm217. PMID 17921483.
  117. ^ Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J (2000). "Chapter 8.5: Gene Replacement and Transgenic Animals: DNA Is Transferred into Eukaryotic Cells in Various Ways". Molecular Cell Biology (4th ed.). W. H. Freeman and Company. ISBN 978-0-7167-3136-8.
  118. ^ "The Nobel Prize in Physiology or Medicine 2007". The Nobel Foundation. Retrieved December 15, 2008.
  119. ^ Drummond DA, Silberg JJ, Meyer MM, Wilke CO, Arnold FH (April 2005). "On the conservative nature of intragenic recombination". Proceedings of the National Academy of Sciences of the United States of America. 102 (15): 5380–5. Bibcode:2005PNAS..102.5380D. doi:10.1073/pnas.0500729102. PMC 556249. PMID 15809422.
  120. ^ Bloom JD, Silberg JJ, Wilke CO, Drummond DA, Adami C, Arnold FH (January 2005). "Thermodynamic prediction of protein neutrality". Proceedings of the National Academy of Sciences of the United States of America. 102 (3): 606–11. arXiv:q-bio/0409013. Bibcode:2005PNAS..102..606B. doi:10.1073/pnas.0406744102. PMC 545518. PMID 15644440.
  121. ^ a b Carbone MN, Arnold FH (August 2007). "Engineering by homologous recombination: exploring sequence and function within a conserved fold". Current Opinion in Structural Biology. 17 (4): 454–9. doi:10.1016/j.sbi.2007.08.005. PMID 17884462.
  122. ^ Otey CR, Landwehr M, Endelman JB, Hiraga K, Bloom JD, Arnold FH (May 2006). "Structure-guided recombination creates an artificial family of cytochromes P450". PLOS Biology. 4 (5): e112. doi:10.1371/journal.pbio.0040112. PMC 1431580. PMID 16594730.  
  123. ^ Socolich M, Lockless SW, Russ WP, Lee H, Gardner KH, Ranganathan R (September 2005). "Evolutionary information for specifying a protein fold". Nature. 437 (7058): 512–8. Bibcode:2005Natur.437..512S. doi:10.1038/nature03991. PMID 16177782. S2CID 4363255.
  124. ^ Thulasiram HV, Erickson HK, Poulter CD (April 2007). "Chimeras of two isoprenoid synthases catalyze all four coupling reactions in isoprenoid biosynthesis". Science. 316 (5821): 73–6. Bibcode:2007Sci...316...73T. doi:10.1126/science.1137786. PMID 17412950. S2CID 43516273.
  125. ^ Landwehr M, Carbone M, Otey CR, Li Y, Arnold FH (March 2007). "Diversification of catalytic function in a synthetic family of chimeric cytochrome p450s". Chemistry & Biology. 14 (3): 269–78. doi:10.1016/j.chembiol.2007.01.009. PMC 1991292. PMID 17379142.
  126. ^ Choudhury, Ananya; Zhao, Helen; Jalali, Farid; Rashid, Shahnaz AL; Ran, Jane; Supiot, Stephane; Kiltie, Anne E.; Bristow, Robert G. (1 January 2009). "Targeting homologous recombination using imatinib results in enhanced tumor cell chemosensitivity and radiosensitivity" (PDF). Molecular Cancer Therapeutics. 8 (1): 203–213. doi:10.1158/1535-7163.MCT-08-0959. ISSN 1535-7163. PMID 19139130. S2CID 6493434.
  127. ^ Siddiqui, Arafat; Tumiati, Manuela; Joko, Alia; Sandholm, Jouko; Roering, Pia; Aakko, Sofia; Vainionpää, Reetta; Kaipio, Katja; Huhtinen, Kaisa; Kauppi, Liisa; Tuomela, Johanna; Hietanen, Sakari (2021). "Targeting DNA Homologous Repair Proficiency With Concomitant Topoisomerase II and c-Abl Inhibition". Frontiers in Oncology. 11: 3666. doi:10.3389/fonc.2021.733700. ISSN 2234-943X. PMC 8488401. PMID 34616682.
  128. ^ a b c Iglehart JD, Silver DP (July 2009). "Synthetic lethality--a new direction in cancer-drug development". The New England Journal of Medicine. 361 (2): 189–91. doi:10.1056/NEJMe0903044. PMID 19553640.
  129. ^ Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ, Ashworth A, Carmichael J, Kaye SB, Schellens JH, de Bono JS (July 2009). "Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers". The New England Journal of Medicine. 361 (2): 123–34. doi:10.1056/NEJMoa0900212. PMID 19553641.
  130. ^ Edwards SL, Brough R, Lord CJ, Natrajan R, Vatcheva R, Levine DA, Boyd J, Reis-Filho JS, Ashworth A (February 2008). "Resistance to therapy caused by intragenic deletion in BRCA2". Nature. 451 (7182): 1111–5. Bibcode:2008Natur.451.1111E. doi:10.1038/nature06548. PMID 18264088. S2CID 205212044.

External links edit

  • Animations – homologous recombination: Animations showing several models of homologous recombination
  • Homologous recombination: Tempy & Trun: Animation of the bacterial RecBCD pathway of homologous recombination

homologous, recombination, type, genetic, recombination, which, genetic, information, exchanged, between, similar, identical, molecules, double, stranded, single, stranded, nucleic, acids, usually, cellular, organisms, also, viruses, figure, during, meiosis, h. Homologous recombination is a type of genetic recombination in which genetic information is exchanged between two similar or identical molecules of double stranded or single stranded nucleic acids usually DNA as in cellular organisms but may be also RNA in viruses Figure 1 During meiosis homologous recombination can produce new combinations of genes as shown here between similar but not identical copies of human chromosome 1 Homologous recombination is widely used by cells to accurately repair harmful DNA breaks that occur on both strands of DNA known as double strand breaks DSB in a process called homologous recombinational repair HRR 1 Homologous recombination also produces new combinations of DNA sequences during meiosis the process by which eukaryotes make gamete cells like sperm and egg cells in animals These new combinations of DNA represent genetic variation in offspring which in turn enables populations to adapt during the course of evolution 2 Homologous recombination is also used in horizontal gene transfer to exchange genetic material between different strains and species of bacteria and viruses Horizontal gene transfer is the primary mechanism for the spread of antibiotic resistance in bacteria Although homologous recombination varies widely among different organisms and cell types for double stranded DNA dsDNA most forms involve the same basic steps After a double strand break occurs sections of DNA around the 5 ends of the break are cut away in a process called resection In the strand invasion step that follows an overhanging 3 end of the broken DNA molecule then invades a similar or identical DNA molecule that is not broken After strand invasion the further sequence of events may follow either of two main pathways discussed below see Models the DSBR double strand break repair pathway or the SDSA synthesis dependent strand annealing pathway Homologous recombination that occurs during DNA repair tends to result in non crossover products in effect restoring the damaged DNA molecule as it existed before the double strand break Homologous recombination is conserved across all three domains of life as well as DNA and RNA viruses suggesting that it is a nearly universal biological mechanism The discovery of genes for homologous recombination in protists a diverse group of eukaryotic microorganisms has been interpreted as evidence that homologous recombination emerged early in the evolution of eukaryotes Since their dysfunction has been strongly associated with increased susceptibility to several types of cancer the proteins that facilitate homologous recombination are topics of active research Homologous recombination is also used in gene targeting a technique for introducing genetic changes into target organisms For their development of this technique Mario Capecchi Martin Evans and Oliver Smithies were awarded the 2007 Nobel Prize for Physiology or Medicine Capecchi 3 and Smithies 4 independently discovered applications to mouse embryonic stem cells however the highly conserved mechanisms underlying the DSB repair model including uniform homologous integration of transformed DNA gene therapy were first shown in plasmid experiments by Orr Weaver Szostak and Rothstein 5 6 7 Researching the plasmid induced DSB using g irradiation 8 in the 1970s 1980s led to later experiments using endonucleases e g I SceI to cut chromosomes for genetic engineering of mammalian cells where nonhomologous recombination is more frequent than in yeast 9 Contents 1 History and discovery 2 In eukaryotes 2 1 Timing within the mitotic cell cycle 2 2 The role of chromatin 2 3 Homologous recombination during meiosis 2 4 Models 2 4 1 DSBR pathway 2 4 2 SDSA pathway 2 4 3 SSA pathway 2 4 4 BIR pathway 3 In bacteria 3 1 RecBCD pathway 3 2 RecF pathway 3 3 Branch migration 3 4 Resolution 3 5 Facilitating genetic transfer 3 6 Bacterial transformation 4 In viruses 4 1 Coronavirus 5 Effects of dysfunction 6 Evolutionary conservation 6 1 The RecA recombinase family 6 2 Meiosis specific proteins 7 Technological applications 7 1 Gene targeting 7 2 Protein engineering 7 3 Cancer therapy 8 See also 9 References 10 External linksHistory and discovery edit nbsp Figure 2 An early illustration of crossing over from Thomas Hunt MorganIn the early 1900s William Bateson and Reginald Punnett found an exception to one of the principles of inheritance originally described by Gregor Mendel in the 1860s In contrast to Mendel s notion that traits are independently assorted when passed from parent to child for example that a cat s hair color and its tail length are inherited independent of each other Bateson and Punnett showed that certain genes associated with physical traits can be inherited together or genetically linked 10 11 In 1911 after observing that linked traits could on occasion be inherited separately Thomas Hunt Morgan suggested that crossovers can occur between linked genes 12 where one of the linked genes physically crosses over to a different chromosome Two decades later Barbara McClintock and Harriet Creighton demonstrated that chromosomal crossover occurs during meiosis 13 14 the process of cell division by which sperm and egg cells are made Within the same year as McClintock s discovery Curt Stern showed that crossing over later called recombination could also occur in somatic cells like white blood cells and skin cells that divide through mitosis 13 15 In 1947 the microbiologist Joshua Lederberg showed that bacteria which had been assumed to reproduce only asexually through binary fission are capable of genetic recombination which is more similar to sexual reproduction This work established E coli as a model organism in genetics 16 and helped Lederberg win the 1958 Nobel Prize in Physiology or Medicine 17 Building on studies in fungi in 1964 Robin Holliday proposed a model for recombination in meiosis which introduced key details of how the process can work including the exchange of material between chromosomes through Holliday junctions 18 In 1983 Jack Szostak and colleagues presented a model now known as the DSBR pathway which accounted for observations not explained by the Holliday model 18 7 During the next decade experiments in Drosophila budding yeast and mammalian cells led to the emergence of other models of homologous recombination called SDSA pathways which do not always rely on Holliday junctions 18 Much of the later work identifying proteins involved in the process and determining their mechanisms has been performed by a number of individuals including James Haber Patrick Sung Stephen Kowalczykowski and others In eukaryotes editHomologous recombination HR is essential to cell division in eukaryotes like plants animals fungi and protists Homologous recombination repairs double strand breaks in DNA caused by ionizing radiation or DNA damaging chemicals 19 Left unrepaired these double strand breaks can cause large scale rearrangement of chromosomes in somatic cells 20 which can in turn lead to cancer 21 In addition to repairing DNA homologous recombination also helps produce genetic diversity when cells divide in meiosis to become specialized gamete cells sperm or egg cells in animals pollen or ovules in plants and spores in fungi It does so by facilitating chromosomal crossover in which regions of similar but not identical DNA are exchanged between homologous chromosomes 22 23 This creates new possibly beneficial combinations of genes which can give offspring an evolutionary advantage 24 Chromosomal crossover often begins when a protein called Spo11 makes a targeted double strand break in DNA 25 These sites are non randomly located on the chromosomes usually in intergenic promoter regions and preferentially in GC rich domains 26 These double strand break sites often occur at recombination hotspots regions in chromosomes that are about 1 000 2 000 base pairs in length and have high rates of recombination The absence of a recombination hotspot between two genes on the same chromosome often means that those genes will be inherited by future generations in equal proportion This represents linkage between the two genes greater than would be expected from genes that independently assort during meiosis 27 Timing within the mitotic cell cycle edit nbsp Figure 3 Homologous recombination repair attempts occur in DNA before the cell enters mitosis M phase shown during the S and G2 phases of the cell cycle Double strand breaks can be repaired through homologous recombination polymerase theta mediated end joining TMEJ or through non homologous end joining NHEJ 28 NHEJ is a DNA repair mechanism which unlike homologous recombination does not require a long homologous sequence to guide repair Whether homologous recombination or NHEJ is used to repair double strand breaks is largely determined by the phase of cell cycle Homologous recombination repairs DNA before the cell enters mitosis M phase It occurs during and shortly after DNA replication in the S and G2 phases of the cell cycle when sister chromatids are more easily available 29 Compared to homologous chromosomes which are similar to another chromosome but often have different alleles sister chromatids are an ideal template for homologous recombination because they are an identical copy of a given chromosome When no homologous template is available or when the template cannot be accessed due to a defect in homologous recombination the break is repaired via TMEJ in the S and G2 phases of the cell cycle In contrast to homologous recombination and TMEJ NHEJ is predominant in the G1 phase of the cell cycle when the cell is growing but not yet ready to divide It occurs less frequently after the G1 phase but maintains at least some activity throughout the cell cycle The mechanisms that regulate homologous recombination and NHEJ throughout the cell cycle vary widely between species 30 Cyclin dependent kinases CDKs which modify the activity of other proteins by adding phosphate groups to that is phosphorylating them are important regulators of homologous recombination in eukaryotes 30 When DNA replication begins in budding yeast the cyclin dependent kinase Cdc28 begins homologous recombination by phosphorylating the Sae2 protein 31 After being so activated by the addition of a phosphate Sae2 causes a clean cut to be made near a double strand break in DNA It is unclear if the endonuclease responsible for this cut is Sae2 itself or another protein Mre11 32 This allows a protein complex including Mre11 known as the MRX complex to bind to DNA and begins a series of protein driven reactions that exchange material between two DNA molecules 33 The role of chromatin edit The packaging of eukaryotic DNA into chromatin presents a barrier to all DNA based processes that require recruitment of enzymes to their sites of action To allow homologous recombination HR DNA repair the chromatin must be remodeled In eukaryotes ATP dependent chromatin remodeling complexes and histone modifying enzymes are two predominant factors employed to accomplish this remodeling process 34 Chromatin relaxation occurs rapidly at the site of a DNA damage 35 In one of the earliest steps the stress activated protein kinase c Jun N terminal kinase JNK phosphorylates SIRT6 on serine 10 in response to double strand breaks or other DNA damage 36 This post translational modification facilitates the mobilization of SIRT6 to DNA damage sites and is required for efficient recruitment of poly ADP ribose polymerase 1 PARP1 to DNA break sites and for efficient repair of DSBs 36 PARP1 protein starts to appear at DNA damage sites in less than a second with half maximum accumulation within 1 6 seconds after the damage occurs 37 Next the chromatin remodeler Alc1 quickly attaches to the product of PARP1 action a poly ADP ribose chain and Alc1 completes arrival at the DNA damage within 10 seconds of the occurrence of the damage 35 About half of the maximum chromatin relaxation presumably due to action of Alc1 occurs by 10 seconds 35 This then allows recruitment of the DNA repair enzyme MRE11 to initiate DNA repair within 13 seconds 37 gH2AX the phosphorylated form of H2AX is also involved in the early steps leading to chromatin decondensation after DNA double strand breaks The histone variant H2AX constitutes about 10 of the H2A histones in human chromatin 38 gH2AX H2AX phosphorylated on serine 139 can be detected as soon as 20 seconds after irradiation of cells with DNA double strand break formation and half maximum accumulation of gH2AX occurs in one minute 38 The extent of chromatin with phosphorylated gH2AX is about two million base pairs at the site of a DNA double strand break 38 gH2AX does not itself cause chromatin decondensation but within 30 seconds of irradiation RNF8 protein can be detected in association with gH2AX 39 RNF8 mediates extensive chromatin decondensation through its subsequent interaction with CHD4 40 a component of the nucleosome remodeling and deacetylase complex NuRD After undergoing relaxation subsequent to DNA damage followed by DNA repair chromatin recovers to a compaction state close to its pre damage level after about 20 min 35 Homologous recombination during meiosis edit In vertebrates the locations at which recombination occurs are determined by the binding locations of PRDM9 a protein which recognizes a specific sequence motif by its zinc finger array 41 At these sites another protein SPO11 catalyses recombination initiating double strand breaks DSBs a subset of which are repaired by recombination with the homologous chromosome PRDM9 deposits both H3K4me3 and H3K36me3 histone methylation marks at the sites it binds and this methyltransferase activity is essential for its role in DSB positioning Following their formation DSB sites are processed by resection resulting in single stranded DNA ssDNA that becomes decorated with DMC1 From mid zygotene to early pachytene as part of the recombinational repair process DMC1 dissociates from the ssDNA and counts decrease until all breaks except those on the XY chromosomes are repaired at late pachytene Several other proteins are involved in this process including ZCWPW1 the 42 first protein directly positioned by PRDM9 s dual histone marks ZCWPW1 is important for homologous DSB repair not positioning Models edit nbsp Figure 4 Double strand break repair models that act via homologous recombinationTwo primary models for how homologous recombination repairs double strand breaks in DNA are the double strand break repair DSBR pathway sometimes called the double Holliday junction model and the synthesis dependent strand annealing SDSA pathway 43 The two pathways are similar in their first several steps After a double strand break occurs the MRX complex MRN complex in humans binds to DNA on either side of the break Next a resection takes place in which DNA around the 5 ends of the break is cut back This happens in two distinct steps first the MRX complex recruits the Sae2 protein and these two proteins trim back the 5 ends on either side of the break to create short 3 overhangs of single strand DNA in the second step 5 3 resection is continued by the Sgs1 helicase and the Exo1 and Dna2 nucleases As a helicase Sgs1 unzips the double strand DNA while the nuclease activity of Exo1 and Dna2 allows them to cut the single stranded DNA produced by Sgs1 31 The RPA protein which has high affinity for single stranded DNA then binds the 3 overhangs 44 With the help of several other proteins that mediate the process the Rad51 protein and Dmc1 in meiosis then forms a filament of nucleic acid and protein on the single strand of DNA coated with RPA This nucleoprotein filament then begins searching for DNA sequences similar to that of the 3 overhang After finding such a sequence the single stranded nucleoprotein filament moves into invades the similar or identical recipient DNA duplex in a process called strand invasion In cells that divide through mitosis the recipient DNA duplex is generally a sister chromatid which is identical to the damaged DNA molecule and provides a template for repair In meiosis however the recipient DNA tends to be from a similar but not necessarily identical homologous chromosome 43 A displacement loop D loop is formed during strand invasion between the invading 3 overhang strand and the homologous chromosome After strand invasion a DNA polymerase extends the end of the invading 3 strand by synthesizing new DNA This changes the D loop to a cross shaped structure known as a Holliday junction Following this more DNA synthesis occurs on the invading strand i e one of the original 3 overhangs effectively restoring the strand on the homologous chromosome that was displaced during strand invasion 43 DSBR pathway edit nbsp Figure 5 The DSBR and SDSA pathways follow the same initial steps but diverge thereafter The DSBR pathway most often results in chromosomal crossover bottom left while SDSA always ends with non crossover products bottom right After the stages of resection strand invasion and DNA synthesis the DSBR and SDSA pathways become distinct 43 The DSBR pathway is unique in that the second 3 overhang which was not involved in strand invasion also forms a Holliday junction with the homologous chromosome The double Holliday junctions are then converted into recombination products by nicking endonucleases a type of restriction endonuclease which cuts only one DNA strand The DSBR pathway commonly results in crossover though it can sometimes result in non crossover products the ability of a broken DNA molecule to collect sequences from separated donor loci was shown in mitotic budding yeast using plasmids or endonuclease induction of chromosomal events 45 46 Because of this tendency for chromosomal crossover the DSBR pathway is a likely model of how crossover homologous recombination occurs during meiosis 22 Whether recombination in the DSBR pathway results in chromosomal crossover is determined by how the double Holliday junction is cut or resolved Chromosomal crossover will occur if one Holliday junction is cut on the crossing strand and the other Holliday junction is cut on the non crossing strand in Figure 5 along the horizontal purple arrowheads at one Holliday junction and along the vertical orange arrowheads at the other Alternatively if the two Holliday junctions are cut on the crossing strands along the horizontal purple arrowheads at both Holliday junctions in Figure 5 then chromosomes without crossover will be produced 47 SDSA pathway edit Homologous recombination via the SDSA pathway occurs in cells that divide through mitosis and meiosis and results in non crossover products In this model the invading 3 strand is extended along the recipient DNA duplex by a DNA polymerase and is released as the Holliday junction between the donor and recipient DNA molecules slides in a process called branch migration The newly synthesized 3 end of the invading strand is then able to anneal to the other 3 overhang in the damaged chromosome through complementary base pairing After the strands anneal a small flap of DNA can sometimes remain Any such flaps are removed and the SDSA pathway finishes with the resealing also known as ligation of any remaining single stranded gaps 48 During mitosis the major homologous recombination pathway for repairing DNA double strand breaks appears to be the SDSA pathway rather than the DSBR pathway 49 The SDSA pathway produces non crossover recombinants Figure 5 During meiosis non crossover recombinants also occur frequently and these appear to arise mainly by the SDSA pathway as well 49 50 Non crossover recombination events occurring during meiosis likely reflect instances of repair of DNA double strand damages or other types of DNA damages SSA pathway edit nbsp Figure 6 Recombination via the SSA pathway occurs between two repeat elements purple on the same DNA duplex and results in deletions of genetic material Click to view animated diagram in Firefox Chrome Safari or Opera web browsers The single strand annealing SSA pathway of homologous recombination repairs double strand breaks between two repeat sequences The SSA pathway is unique in that it does not require a separate similar or identical molecule of DNA like the DSBR or SDSA pathways of homologous recombination Instead the SSA pathway only requires a single DNA duplex and uses the repeat sequences as the identical sequences that homologous recombination needs for repair The pathway is relatively simple in concept after two strands of the same DNA duplex are cut back around the site of the double strand break the two resulting 3 overhangs then align and anneal to each other restoring the DNA as a continuous duplex 48 51 As DNA around the double strand break is cut back the single stranded 3 overhangs being produced are coated with the RPA protein which prevents the 3 overhangs from sticking to themselves 52 A protein called Rad52 then binds each of the repeat sequences on either side of the break and aligns them to enable the two complementary repeat sequences to anneal 52 After annealing is complete leftover non homologous flaps of the 3 overhangs are cut away by a set of nucleases known as Rad1 Rad10 which are brought to the flaps by the Saw1 and Slx4 proteins 52 53 New DNA synthesis fills in any gaps and ligation restores the DNA duplex as two continuous strands 54 The DNA sequence between the repeats is always lost as is one of the two repeats The SSA pathway is considered mutagenic since it results in such deletions of genetic material 48 BIR pathway edit During DNA replication double strand breaks can sometimes be encountered at replication forks as DNA helicase unzips the template strand These defects are repaired in the break induced replication BIR pathway of homologous recombination The precise molecular mechanisms of the BIR pathway remain unclear Three proposed mechanisms have strand invasion as an initial step but they differ in how they model the migration of the D loop and later phases of recombination 55 The BIR pathway can also help to maintain the length of telomeres regions of DNA at the end of eukaryotic chromosomes in the absence of or in cooperation with telomerase Without working copies of the enzyme telomerase telomeres typically shorten with each cycle of mitosis which eventually blocks cell division and leads to senescence In budding yeast cells where telomerase has been inactivated through mutations two types of survivor cells have been observed to avoid senescence longer than expected by elongating their telomeres through BIR pathways 55 Maintaining telomere length is critical for cell immortalization a key feature of cancer Most cancers maintain telomeres by upregulating telomerase However in several types of human cancer a BIR like pathway helps to sustain some tumors by acting as an alternative mechanism of telomere maintenance 56 This fact has led scientists to investigate whether such recombination based mechanisms of telomere maintenance could thwart anti cancer drugs like telomerase inhibitors 57 In bacteria edit nbsp Figure 7 Crystal structure of a RecA protein filament bound to DNA 58 A 3 overhang is visible to the right of center Homologous recombination is a major DNA repair process in bacteria It is also important for producing genetic diversity in bacterial populations although the process differs substantially from meiotic recombination which repairs DNA damages and brings about diversity in eukaryotic genomes Homologous recombination has been most studied and is best understood for Escherichia coli 59 Double strand DNA breaks in bacteria are repaired by the RecBCD pathway of homologous recombination Breaks that occur on only one of the two DNA strands known as single strand gaps are thought to be repaired by the RecF pathway 60 Both the RecBCD and RecF pathways include a series of reactions known as branch migration in which single DNA strands are exchanged between two intercrossed molecules of duplex DNA and resolution in which those two intercrossed molecules of DNA are cut apart and restored to their normal double stranded state RecBCD pathway edit nbsp Figure 8A Molecular model for the RecBCD pathway of recombination This model is based on reactions of DNA and RecBCD with ATP in excess over Mg2 ions Step 1 RecBCD binds to a double stranded DNA end Step 2 RecBCD unwinds DNA RecD is a fast helicase on the 5 ended strand and RecB is a slower helicase on the 3 ended strand that with an arrowhead ref 46 in current Wiki version This produces two single stranded ss DNA tails and one ss loop The loop and tails enlarge as RecBCD moves along the DNA Step 3 The two tails anneal to produce a second ss DNA loop and both loops move and grow Step 4 Upon reaching the Chi hotspot sequence 5 GCTGGTGG 3 red dot RecBCD nicks the 3 ended strand Further unwinding produces a long 3 ended ss tail with Chi near its end Step 5 RecBCD loads RecA protein onto the Chi tail At some undetermined point the RecBCD subunits disassemble Step 6 The RecA ssDNA complex invades an intact homologous duplex DNA to produce a D loop which can be resolved into intact recombinant DNA in two ways Step 7 The D loop is cut and anneals with the gap in the first DNA to produce a Holliday junction Resolution of the Holliday junction cutting swapping of strands and ligation at the open arrowheads by some combination of RuvABC and RecG produces two recombinants of reciprocal type Step 8 The 3 end of the Chi tail primes DNA synthesis from which a replication fork can be generated Resolution of the fork at the open arrowheads produces one recombinant non reciprocal DNA one parental type DNA and one DNA fragment 61 nbsp Figure 8B Beginning of the RecBCD pathway This model is based on reactions of DNA and RecBCD with Mg2 ions in excess over ATP Step 1 RecBCD binds to a DNA double strand break Step 2 RecBCD initiates unwinding of the DNA duplex through ATP dependent helicase activity Step 3 RecBCD continues its unwinding and moves down the DNA duplex cleaving the 3 strand much more frequently than the 5 strand Step 4 RecBCD encounters a Chi sequence and stops digesting the 3 strand cleavage of the 5 strand is significantly increased Step 5 RecBCD loads RecA onto the 3 strand Step 6 RecBCD unbinds from the DNA duplex leaving a RecA nucleoprotein filament on the 3 tail 62 The RecBCD pathway is the main recombination pathway used in many bacteria to repair double strand breaks in DNA and the proteins are found in a broad array of bacteria 63 64 65 These double strand breaks can be caused by UV light and other radiation as well as chemical mutagens Double strand breaks may also arise by DNA replication through a single strand nick or gap Such a situation causes what is known as a collapsed replication fork and is fixed by several pathways of homologous recombination including the RecBCD pathway 66 In this pathway a three subunit enzyme complex called RecBCD initiates recombination by binding to a blunt or nearly blunt end of a break in double strand DNA After RecBCD binds the DNA end the RecB and RecD subunits begin unzipping the DNA duplex through helicase activity The RecB subunit also has a nuclease domain which cuts the single strand of DNA that emerges from the unzipping process This unzipping continues until RecBCD encounters a specific nucleotide sequence 5 GCTGGTGG 3 known as a Chi site 65 Upon encountering a Chi site the activity of the RecBCD enzyme changes drastically 64 61 67 DNA unwinding pauses for a few seconds and then resumes at roughly half the initial speed This is likely because the slower RecB helicase unwinds the DNA after Chi rather than the faster RecD helicase which unwinds the DNA before Chi 68 69 Recognition of the Chi site also changes the RecBCD enzyme so that it cuts the DNA strand with Chi and begins loading multiple RecA proteins onto the single stranded DNA with the newly generated 3 end The resulting RecA coated nucleoprotein filament then searches out similar sequences of DNA on a homologous chromosome The search process induces stretching of the DNA duplex which enhances homology recognition a mechanism termed conformational proofreading 70 71 72 Upon finding such a sequence the single stranded nucleoprotein filament moves into the homologous recipient DNA duplex in a process called strand invasion 73 The invading 3 overhang causes one of the strands of the recipient DNA duplex to be displaced to form a D loop If the D loop is cut another swapping of strands forms a cross shaped structure called a Holliday junction 65 Resolution of the Holliday junction by some combination of RuvABC or RecG can produce two recombinant DNA molecules with reciprocal genetic types if the two interacting DNA molecules differ genetically Alternatively the invading 3 end near Chi can prime DNA synthesis and form a replication fork This type of resolution produces only one type of recombinant non reciprocal RecF pathway edit Further information RecF pathway Bacteria appear to use the RecF pathway of homologous recombination to repair single strand gaps in DNA When the RecBCD pathway is inactivated by mutations and additional mutations inactivate the SbcCD and ExoI nucleases the RecF pathway can also repair DNA double strand breaks 74 In the RecF pathway the RecQ helicase unwinds the DNA and the RecJ nuclease degrades the strand with a 5 end leaving the strand with the 3 end intact RecA protein binds to this strand and is either aided by the RecF RecO and RecR proteins or stabilized by them The RecA nucleoprotein filament then searches for a homologous DNA and exchanges places with the identical or nearly identical strand in the homologous DNA Although the proteins and specific mechanisms involved in their initial phases differ the two pathways are similar in that they both require single stranded DNA with a 3 end and the RecA protein for strand invasion The pathways are also similar in their phases of branch migration in which the Holliday junction slides in one direction and resolution in which the Holliday junctions are cleaved apart by enzymes 75 76 The alternative non reciprocal type of resolution may also occur by either pathway Branch migration edit Immediately after strand invasion the Holliday junction moves along the linked DNA during the branch migration process It is in this movement of the Holliday junction that base pairs between the two homologous DNA duplexes are exchanged To catalyze branch migration the RuvA protein first recognizes and binds to the Holliday junction and recruits the RuvB protein to form the RuvAB complex Two sets of the RuvB protein which each form a ring shaped ATPase are loaded onto opposite sides of the Holliday junction where they act as twin pumps that provide the force for branch migration Between those two rings of RuvB two sets of the RuvA protein assemble in the center of the Holliday junction such that the DNA at the junction is sandwiched between each set of RuvA The strands of both DNA duplexes the donor and the recipient duplexes are unwound on the surface of RuvA as they are guided by the protein from one duplex to the other 77 78 Resolution edit In the resolution phase of recombination any Holliday junctions formed by the strand invasion process are cut thereby restoring two separate DNA molecules This cleavage is done by RuvAB complex interacting with RuvC which together form the RuvABC complex RuvC is an endonuclease that cuts the degenerate sequence 5 A T TT G C 3 The sequence is found frequently in DNA about once every 64 nucleotides 78 Before cutting RuvC likely gains access to the Holliday junction by displacing one of the two RuvA tetramers covering the DNA there 77 Recombination results in either splice or patch products depending on how RuvC cleaves the Holliday junction 78 Splice products are crossover products in which there is a rearrangement of genetic material around the site of recombination Patch products on the other hand are non crossover products in which there is no such rearrangement and there is only a patch of hybrid DNA in the recombination product 79 Facilitating genetic transfer edit Homologous recombination is an important method of integrating donor DNA into a recipient organism s genome in horizontal gene transfer the process by which an organism incorporates foreign DNA from another organism without being the offspring of that organism Homologous recombination requires incoming DNA to be highly similar to the recipient genome and so horizontal gene transfer is usually limited to similar bacteria 80 Studies in several species of bacteria have established that there is a log linear decrease in recombination frequency with increasing difference in sequence between host and recipient DNA 81 82 83 In bacterial conjugation where DNA is transferred between bacteria through direct cell to cell contact homologous recombination helps integrate foreign DNA into the host genome via the RecBCD pathway The RecBCD enzyme promotes recombination after DNA is converted from single strand DNA in which form it originally enters the bacterium to double strand DNA during replication The RecBCD pathway is also essential for the final phase of transduction a type of horizontal gene transfer in which DNA is transferred from one bacterium to another by a virus Foreign bacterial DNA is sometimes misincorporated in the capsid head of bacteriophage virus particles as DNA is packaged into new bacteriophages during viral replication When these new bacteriophages infect other bacteria DNA from the previous host bacterium is injected into the new bacterial host as double strand DNA The RecBCD enzyme then incorporates this double strand DNA into the genome of the new bacterial host 65 Bacterial transformation edit Natural bacterial transformation involves the transfer of DNA from a donor bacterium to a recipient bacterium where both donor and recipient are ordinarily of the same species Transformation unlike bacterial conjugation and transduction depends on numerous bacterial gene products that specifically interact to perform this process 84 Thus transformation is clearly a bacterial adaptation for DNA transfer In order for a bacterium to bind take up and integrate donor DNA into its resident chromosome by homologous recombination it must first enter a special physiological state termed competence The RecA Rad51 DMC1 gene family plays a central role in homologous recombination during bacterial transformation as it does during eukaryotic meiosis and mitosis For instance the RecA protein is essential for transformation in Bacillus subtilis and Streptococcus pneumoniae 85 and expression of the RecA gene is induced during the development of competence for transformation in these organisms As part of the transformation process the RecA protein interacts with entering single stranded DNA ssDNA to form RecA ssDNA nucleofilaments that scan the resident chromosome for regions of homology and bring the entering ssDNA to the corresponding region where strand exchange and homologous recombination occur 86 Thus the process of homologous recombination during bacterial transformation has fundamental similarities to homologous recombination during meiosis In viruses editHomologous recombination occurs in several groups of viruses In DNA viruses such as herpesvirus recombination occurs through a break and rejoin mechanism like in bacteria and eukaryotes 87 There is also evidence for recombination in some RNA viruses specifically positive sense ssRNA viruses like retroviruses picornaviruses and coronaviruses There is controversy over whether homologous recombination occurs in negative sense ssRNA viruses like influenza 88 In RNA viruses homologous recombination can be either precise or imprecise In the precise type of RNA RNA recombination there is no difference between the two parental RNA sequences and the resulting crossover RNA region Because of this it is often difficult to determine the location of crossover events between two recombining RNA sequences In imprecise RNA homologous recombination the crossover region has some difference with the parental RNA sequences caused by either addition deletion or other modification of nucleotides The level of precision in crossover is controlled by the sequence context of the two recombining strands of RNA sequences rich in adenine and uracil decrease crossover precision 89 90 Homologous recombination is important in facilitating viral evolution 89 91 For example if the genomes of two viruses with different disadvantageous mutations undergo recombination then they may be able to regenerate a fully functional genome Alternatively if two similar viruses have infected the same host cell homologous recombination can allow those two viruses to swap genes and thereby evolve more potent variations of themselves 91 Homologous recombination is the proposed mechanism whereby the DNA virus human herpesvirus 6 integrates into human telomeres 92 When two or more viruses each containing lethal genomic damage infect the same host cell the virus genomes can often pair with each other and undergo homologous recombinational repair to produce viable progeny This process known as multiplicity reactivation has been studied in several bacteriophages including phage T4 93 Enzymes employed in recombinational repair in phage T4 are functionally homologous to enzymes employed in bacterial and eukaryotic recombinational repair 94 In particular with regard to a gene necessary for the strand exchange reaction a key step in homologous recombinational repair there is functional homology from viruses to humans i e uvsX in phage T4 recA in E coli and other bacteria and rad51 and dmc1 in yeast and other eukaryotes including humans 95 Multiplicity reactivation has also been demonstrated in numerous pathogenic viruses 96 Coronavirus edit Coronaviruses are capable of genetic recombination when at least two viral genomes are present in the same infected cell RNA recombination appears to be a major driving force in determining 1 genetic variability within a CoV species 2 the capability of a CoV species to jump from one host to another and 3 infrequently the emergence of novel CoVs 97 The mechanism of recombination in CoVs likely involves template switching during genome replication 97 Recombination in RNA viruses appears to be an adaptation for coping with genome damage 98 The pandemic SARS CoV 2 s entire receptor binding motif appears to have been introduced through recombination from coronaviruses of pangolins 99 Such a recombination event may have been a critical step in the evolution of SARS CoV 2 s capability to infect humans 99 Recombination events are likely key steps in the evolutionary process that leads to the emergence of new human coronaviruses 100 During COVID 19 pandemic in 2020 many genomic sequences of Australian SARS CoV 2 isolates have deletions or mutations 29742G gt A or 29742G gt U G19A or G19U in the Coronavirus 3 stem loop II like motif s2m an RNA motif in 3 untranslated region of viral genome suggesting that RNA recombination events may have occurred in s2m of SARS CoV 2 Based on computational analysis of 1319 Australia SARS CoV 2 sequences using Recco algorithm https recco bioinf mpi inf mpg de 29742G G19 29744G G21 and 29751G G28 were predicted as recombination hotspots 101 nbsp Schematic representation of the s2m RNA secondary structure with tertiary structural interactions indicated as long range contacts The SARS CoV 2 outbreak in Diamond Princess cruise most likely originated from either a single person infected with a virus variant identical to the Wuhan WIV04 isolates or simultaneously with another primary case infected with a virus containing the 11083G gt T mutation Linkage disequilibrium analysis confirmed that RNA recombination with the 11083G gt T mutation also contributed to the increase of mutations among the viral progeny The findings indicate that the 11083G gt T mutation of SARS CoV 2 spread during shipboard quarantine and arose through de novo RNA recombination under positive selection pressure In addition in three patients in this cruise two mutations 29736G gt T and 29751G gt T G13 and G28 were also located in Coronavirus 3 stem loop II like motif s2m as G28 was predicted as recombination hotspots in Australian SARS CoV 2 mutants Although s2m is considered an RNA motif highly conserved among many coronavirus species this result also suggests that s2m of SARS CoV 2 is rather a RNA recombination mutation hotspot 102 Effects of dysfunction edit nbsp Figure 9 Joining of single ended double strand breaks could lead to rearrangementsWithout proper homologous recombination chromosomes often incorrectly align for the first phase of cell division in meiosis This causes chromosomes to fail to properly segregate in a process called nondisjunction In turn nondisjunction can cause sperm and ova to have too few or too many chromosomes Down s syndrome which is caused by an extra copy of chromosome 21 is one of many abnormalities that result from such a failure of homologous recombination in meiosis 78 103 Deficiencies in homologous recombination have been strongly linked to cancer formation in humans For example each of the cancer related diseases Bloom syndrome Werner syndrome and Rothmund Thomson syndrome are caused by malfunctioning copies of RecQ helicase genes involved in the regulation of homologous recombination BLM WRN and RECQL4 respectively 104 In the cells of Bloom s syndrome patients who lack a working copy of the BLM protein there is an elevated rate of homologous recombination 105 Experiments in mice deficient in BLM have suggested that the mutation gives rise to cancer through a loss of heterozygosity caused by increased homologous recombination 106 A loss in heterozygosity refers to the loss of one of two versions or alleles of a gene If one of the lost alleles helps to suppress tumors like the gene for the retinoblastoma protein for example then the loss of heterozygosity can lead to cancer 107 1236 Decreased rates of homologous recombination cause inefficient DNA repair 107 310 which can also lead to cancer 108 This is the case with BRCA1 and BRCA2 two similar tumor suppressor genes whose malfunctioning has been linked with considerably increased risk for breast and ovarian cancer Cells missing BRCA1 and BRCA2 have a decreased rate of homologous recombination and increased sensitivity to ionizing radiation suggesting that decreased homologous recombination leads to increased susceptibility to cancer 108 Because the only known function of BRCA2 is to help initiate homologous recombination researchers have speculated that more detailed knowledge of BRCA2 s role in homologous recombination may be the key to understanding the causes of breast and ovarian cancer 108 Tumours with a homologous recombination deficiency including BRCA defects are described as HRD positive 109 Evolutionary conservation edit nbsp Figure 10 Protein domains in homologous recombination related proteins are conserved across the three main groups of life archaea bacteria and eukaryotes While the pathways can mechanistically vary the ability of organisms to perform homologous recombination is universally conserved across all domains of life 110 Based on the similarity of their amino acid sequences homologs of a number of proteins can be found in multiple domains of life indicating that they evolved a long time ago and have since diverged from common ancestral proteins 110 RecA recombinase family members are found in almost all organisms with RecA in bacteria Rad51 and DMC1 in eukaryotes RadA in archaea and UvsX in T4 phage 111 Related single stranded binding proteins that are important for homologous recombination and many other processes are also found in all domains of life 112 Rad54 Mre11 Rad50 and a number of other proteins are also found in both archaea and eukaryotes 110 111 113 The RecA recombinase family edit The proteins of the RecA recombinase family of proteins are thought to be descended from a common ancestral recombinase 110 The RecA recombinase family contains RecA protein from bacteria the Rad51 and Dmc1 proteins from eukaryotes and RadA from archaea and the recombinase paralog proteins Studies modeling the evolutionary relationships between the Rad51 Dmc1 and RadA proteins indicate that they are monophyletic or that they share a common molecular ancestor 110 Within this protein family Rad51 and Dmc1 are grouped together in a separate clade from RadA One of the reasons for grouping these three proteins together is that they all possess a modified helix turn helix motif which helps the proteins bind to DNA toward their N terminal ends 110 An ancient gene duplication event of a eukaryotic RecA gene and subsequent mutation has been proposed as a likely origin of the modern RAD51 and DMC1 genes 110 The proteins generally share a long conserved region known as the RecA Rad51 domain Within this protein domain are two sequence motifs Walker A motif and Walker B motif The Walker A and B motifs allow members of the RecA Rad51 protein family to engage in ATP binding and ATP hydrolysis 110 114 Meiosis specific proteins edit The discovery of Dmc1 in several species of Giardia one of the earliest protists to diverge as a eukaryote suggests that meiotic homologous recombination and thus meiosis itself emerged very early in eukaryotic evolution 115 In addition to research on Dmc1 studies on the Spo11 protein have provided information on the origins of meiotic recombination 116 Spo11 a type II topoisomerase can initiate homologous recombination in meiosis by making targeted double strand breaks in DNA 25 Phylogenetic trees based on the sequence of genes similar to SPO11 in animals fungi plants protists and archaea have led scientists to believe that the version Spo11 currently in eukaryotes emerged in the last common ancestor of eukaryotes and archaea 116 Technological applications editGene targeting edit nbsp Figure 11 As a developing embryo this chimeric mouse had the agouti coat color gene introduced into its DNA via gene targeting Its offspring are homozygous for the agouti gene Main article Gene targeting Many methods for introducing DNA sequences into organisms to create recombinant DNA and genetically modified organisms use the process of homologous recombination 117 Also called gene targeting the method is especially common in yeast and mouse genetics The gene targeting method in knockout mice uses mouse embryonic stem cells to deliver artificial genetic material mostly of therapeutic interest which represses the target gene of the mouse by the principle of homologous recombination The mouse thereby acts as a working model to understand the effects of a specific mammalian gene In recognition of their discovery of how homologous recombination can be used to introduce genetic modifications in mice through embryonic stem cells Mario Capecchi Martin Evans and Oliver Smithies were awarded the 2007 Nobel Prize for Physiology or Medicine 118 Advances in gene targeting technologies which hijack the homologous recombination mechanics of cells are now leading to the development of a new wave of more accurate isogenic human disease models These engineered human cell models are thought to more accurately reflect the genetics of human diseases than their mouse model predecessors This is largely because mutations of interest are introduced into endogenous genes just as they occur in the real patients and because they are based on human genomes rather than rat genomes Furthermore certain technologies enable the knock in of a particular mutation rather than just knock outs associated with older gene targeting technologies Protein engineering edit Protein engineering with homologous recombination develops chimeric proteins by swapping fragments between two parental proteins These techniques exploit the fact that recombination can introduce a high degree of sequence diversity while preserving a protein s ability to fold into its tertiary structure or three dimensional shape 119 This stands in contrast to other protein engineering techniques like random point mutagenesis in which the probability of maintaining protein function declines exponentially with increasing amino acid substitutions 120 The chimeras produced by recombination techniques are able to maintain their ability to fold because their swapped parental fragments are structurally and evolutionarily conserved These recombinable building blocks preserve structurally important interactions like points of physical contact between different amino acids in the protein s structure Computational methods like SCHEMA and statistical coupling analysis can be used to identify structural subunits suitable for recombination 121 122 123 Techniques that rely on homologous recombination have been used to engineer new proteins 121 In a study published in 2007 researchers were able to create chimeras of two enzymes involved in the biosynthesis of isoprenoids a diverse class of compounds including hormones visual pigments and certain pheromones The chimeric proteins acquired an ability to catalyze an essential reaction in isoprenoid biosynthesis one of the most diverse pathways of biosynthesis found in nature that was absent in the parent proteins 124 Protein engineering through recombination has also produced chimeric enzymes with new function in members of a group of proteins known as the cytochrome P450 family 125 which in humans is involved in detoxifying foreign compounds like drugs food additives and preservatives 22 Cancer therapy edit Homologous recombination proficient HRP cancer cells are able to repair the DNA damage which is caused by chemotherapy such as cisplatin Thus HRP cancers are difficult to treat Studies suggest that homologous recombination can be targeted via c Abl inhibition 126 127 Cancer cells with BRCA mutations have deficiencies in homologous recombination and drugs to exploit those deficiencies have been developed and used successfully in clinical trials 128 129 Olaparib a PARP1 inhibitor shrunk or stopped the growth of tumors from breast ovarian and prostate cancers caused by mutations in the BRCA1 or BRCA2 genes which are necessary for HR When BRCA1 or BRCA2 is absent other types of DNA repair mechanisms must compensate for the deficiency of HR such as base excision repair BER for stalled replication forks or non homologous end joining NHEJ for double strand breaks 128 By inhibiting BER in an HR deficient cell olaparib applies the concept of synthetic lethality to specifically target cancer cells While PARP1 inhibitors represent a novel approach to cancer therapy researchers have cautioned that they may prove insufficient for treating late stage metastatic cancers 128 Cancer cells can become resistant to a PARP1 inhibitor if they undergo deletions of mutations in BRCA2 undermining the drug s synthetic lethality by restoring cancer cells ability to repair DNA by HR 130 See also editChromosomal crossover Homology directed repairReferences edit Thompson LH Schild D June 2001 Homologous recombinational repair of DNA ensures mammalian chromosome stability Mutation Research 477 1 2 131 53 doi 10 1016 S0027 5107 01 00115 4 PMID 11376695 Alberts B Johnson A Lewis J Raff M Roberts K Walter P et al 2002 Chapter 5 DNA Replication Repair and Recombination Molecular Biology of the Cell 4th ed New York Garland Science p 845 ISBN 978 0 8153 3218 3 OCLC 145080076 Capecchi MR June 1989 Altering the genome by homologous recombination Science 244 4910 1288 92 Bibcode 1989Sci 244 1288C doi 10 1126 science 2660260 PMID 2660260 Smithies O Gregg RG Boggs SS Koralewski MA Kucherlapati RS 1985 09 19 Insertion of DNA sequences into the human chromosomal beta globin locus by homologous recombination Nature 317 6034 230 4 Bibcode 1985Natur 317 230S doi 10 1038 317230a0 PMID 2995814 S2CID 30212766 Orr Weaver TL Szostak JW Rothstein RJ October 1981 Yeast transformation a model system for the study of recombination Proceedings of the National Academy of Sciences of the United States of America 78 10 6354 8 Bibcode 1981PNAS 78 6354O doi 10 1073 pnas 78 10 6354 PMC 349037 PMID 6273866 Orr Weaver TL Szostak JW July 1983 Yeast recombination the association between double strand gap repair and crossing over Proceedings of the National Academy of Sciences of the United States of America 80 14 4417 21 Bibcode 1983PNAS 80 4417O doi 10 1073 pnas 80 14 4417 PMC 384049 PMID 6308623 a b Szostak JW Orr Weaver TL Rothstein RJ Stahl FW May 1983 The double strand break repair model for recombination Cell 33 1 25 35 doi 10 1016 0092 8674 83 90331 8 PMID 6380756 S2CID 39590123 Resnick MA June 1976 The repair of double strand breaks in DNA a model involving recombination Journal of Theoretical Biology 59 1 97 106 Bibcode 1976JThBi 59 97R doi 10 1016 s0022 5193 76 80025 2 PMID 940351 Jasin M Rothstein R November 2013 Repair of strand breaks by homologous recombination Cold Spring Harbor Perspectives in Biology 5 11 a012740 doi 10 1101 cshperspect a012740 PMC 3809576 PMID 24097900 Bateson P August 2002 William Bateson a biologist ahead of his time PDF Journal of Genetics 81 2 49 58 doi 10 1007 BF02715900 PMID 12532036 S2CID 26806110 Reginald Crundall Punnett NAHSTE University of Edinburgh Retrieved 3 July 2010 Lobo I Shaw K 2008 Thomas Hunt Morgan genetic recombination and gene mapping Nature Education 1 1 a b Coe E Kass LB May 2005 Proof of physical exchange of genes on the chromosomes Proceedings of the National Academy of Sciences of the United States of America 102 19 6641 6 Bibcode 2005PNAS 102 6641C doi 10 1073 pnas 0407340102 PMC 1100733 PMID 15867161 Creighton HB McClintock B August 1931 A Correlation of Cytological and Genetical Crossing Over in Zea Mays Proceedings of the National Academy of Sciences of the United States of America 17 8 492 7 Bibcode 1931PNAS 17 492C doi 10 1073 pnas 17 8 492 PMC 1076098 PMID 16587654 Stern C 1931 Zytologisch genetische untersuchungen alsbeweise fur die Morgansche theorie des faktoraustauschs Biologisches Zentralblatt 51 547 587 The development of bacterial genetics US National Library of Medicine Retrieved 3 July 2010 The Nobel Prize in Physiology or Medicine 1958 Nobelprize org Retrieved 3 July 2010 a b c Haber JE Ira G Malkova A Sugawara N January 2004 Repairing a double strand chromosome break by homologous recombination revisiting Robin Holliday s model Philosophical Transactions of the Royal Society of London Series B Biological Sciences 359 1441 79 86 doi 10 1098 rstb 2003 1367 PMC 1693306 PMID 15065659 Lodish H Berk A Zipursky SL Matsudaira P Baltimore D Darnell J 2000 12 5 Recombination between Homologous DNA Sites Double Strand Breaks in DNA Initiate Recombination Molecular Cell Biology 4th ed W H Freeman and Company ISBN 978 0 7167 3136 8 Griffiths A et al 1999 8 Chromosome Mutations Chromosomal Rearrangements Modern Genetic Analysis W H Freeman and Company ISBN 978 0 7167 3118 4 Khanna KK Jackson SP March 2001 DNA double strand breaks signaling repair and the cancer connection Nature Genetics 27 3 247 54 doi 10 1038 85798 PMID 11242102 S2CID 3012823 a b c Nelson DL Cox MM 2005 Principles of Biochemistry 4th ed Freeman pp 980 981 ISBN 978 0 7167 4339 2 Marcon E Moens PB August 2005 The evolution of meiosis recruitment and modification of somatic DNA repair proteins BioEssays 27 8 795 808 doi 10 1002 bies 20264 PMID 16015600 S2CID 27658497 Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2008 Molecular Biology of the Cell 5th ed Garland Science p 305 ISBN 978 0 8153 4105 5 a b Keeney S Giroux CN Kleckner N February 1997 Meiosis specific DNA double strand breaks are catalyzed by Spo11 a member of a widely conserved protein family Cell 88 3 375 84 doi 10 1016 S0092 8674 00 81876 0 PMID 9039264 S2CID 8294596 Longhese MP Bonetti D Guerini I Manfrini N Clerici M September 2009 DNA double strand breaks in meiosis checking their formation processing and repair DNA Repair 8 9 1127 38 doi 10 1016 j dnarep 2009 04 005 PMID 19464965 Cahill LP Mariana JC Mauleon P January 1979 Total follicular populations in ewes of high and low ovulation rates Journal of Reproduction and Fertility 55 1 27 36 doi 10 1530 jrf 0 0550027 PMID 423159 Schimmel J van Schendel R den Dunnen JT Tijsterman M September 2019 Templated Insertions A Smoking Gun for Polymerase Theta Mediated End Joining Trends in Genetics 35 9 632 644 doi 10 1016 j tig 2019 06 001 PMID 31296341 S2CID 195892718 Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2008 Molecular Biology of the Cell 5th ed Garland Science p 303 ISBN 978 0 8153 4105 5 a b Shrivastav M De Haro LP Nickoloff JA January 2008 Regulation of DNA double strand break repair pathway choice Cell Research 18 1 134 47 doi 10 1038 cr 2007 111 PMID 18157161 a b Mimitou EP Symington LS May 2009 Nucleases and helicases take center stage in homologous recombination Trends in Biochemical Sciences 34 5 264 72 doi 10 1016 j tibs 2009 01 010 PMID 19375328 Andres Sara N Williams R Scott August 2017 CtIP Ctp1 Sae2 molecular form fit for function DNA Repair 56 109 117 doi 10 1016 j dnarep 2017 06 013 PMC 5543718 PMID 28623092 Huertas P Cortes Ledesma F Sartori AA Aguilera A Jackson SP October 2008 CDK targets Sae2 to control DNA end resection and homologous recombination Nature 455 7213 689 92 Bibcode 2008Natur 455 689H doi 10 1038 nature07215 PMC 2635538 PMID 18716619 Liu B Yip RK Zhou Z 2012 Chromatin remodeling DNA damage repair and aging Curr Genomics 13 7 533 47 doi 10 2174 138920212803251373 PMC 3468886 PMID 23633913 a b c d Sellou H Lebeaupin T Chapuis C Smith R Hegele A Singh HR Kozlowski M Bultmann S Ladurner AG Timinszky G Huet S 2016 The poly ADP ribose dependent chromatin remodeler Alc1 induces local chromatin relaxation upon DNA damage Mol Biol Cell 27 24 3791 3799 doi 10 1091 mbc E16 05 0269 PMC 5170603 PMID 27733626 a b Van Meter M Simon M Tombline G May A Morello TD Hubbard BP Bredbenner K Park R Sinclair DA Bohr VA Gorbunova V Seluanov A 2016 JNK Phosphorylates SIRT6 to Stimulate DNA Double Strand Break Repair in Response to Oxidative Stress by Recruiting PARP1 to DNA Breaks Cell Rep 16 10 2641 50 doi 10 1016 j celrep 2016 08 006 PMC 5089070 PMID 27568560 a b Haince JF McDonald D Rodrigue A Dery U Masson JY Hendzel MJ Poirier GG 2008 PARP1 dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites J Biol Chem 283 2 1197 208 doi 10 1074 jbc M706734200 PMID 18025084 a b c Rogakou EP Pilch DR Orr AH Ivanova VS Bonner WM 1998 DNA double stranded breaks induce histone H2AX phosphorylation on serine 139 J Biol Chem 273 10 5858 68 doi 10 1074 jbc 273 10 5858 PMID 9488723 Mailand N Bekker Jensen S Faustrup H Melander F Bartek J Lukas C Lukas J 2007 RNF8 ubiquitylates histones at DNA double strand breaks and promotes assembly of repair proteins Cell 131 5 887 900 doi 10 1016 j cell 2007 09 040 PMID 18001824 S2CID 14232192 Luijsterburg MS Acs K Ackermann L Wiegant WW Bekker Jensen S Larsen DH Khanna KK van Attikum H Mailand N Dantuma NP 2012 A new non catalytic role for ubiquitin ligase RNF8 in unfolding higher order chromatin structure EMBO J 31 11 2511 27 doi 10 1038 emboj 2012 104 PMC 3365417 PMID 22531782 Baudat F Buard J Grey C Fledel Alon A Ober C Przeworski M et al February 2010 PRDM9 is a major determinant of meiotic recombination hotspots in humans and mice Science 327 5967 836 40 Bibcode 2010Sci 327 836B doi 10 1126 science 1183439 PMC 4295902 PMID 20044539 Wells D Bitoun E Moralli D Zhang G Hinch A Jankowska J et al August 2020 ZCWPW1 is recruited to recombination hotspots by PRDM9 and is essential for meiotic double strand break repair eLife 9 e53392 doi 10 7554 eLife 53392 PMC 7494361 PMID 32744506 a b c d Sung P Klein H October 2006 Mechanism of homologous recombination mediators and helicases take on regulatory functions Nature Reviews Molecular Cell Biology 7 10 739 50 doi 10 1038 nrm2008 PMID 16926856 S2CID 30324005 Wold MS 1997 Replication protein A a heterotrimeric single stranded DNA binding protein required for eukaryotic DNA metabolism Annual Review of Biochemistry 66 61 92 doi 10 1146 annurev biochem 66 1 61 PMID 9242902 McMahill MS Sham CW Bishop DK November 2007 Synthesis dependent strand annealing in meiosis PLOS Biology 5 11 e299 doi 10 1371 journal pbio 0050299 PMC 2062477 PMID 17988174 nbsp Bartsch S Kang LE Symington LS February 2000 RAD51 is required for the repair of plasmid double stranded DNA gaps from either plasmid or chromosomal templates Molecular and Cellular Biology 20 4 1194 205 doi 10 1128 MCB 20 4 1194 1205 2000 PMC 85244 PMID 10648605 Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2008 Molecular Biology of the Cell 5th ed Garland Science pp 312 313 ISBN 978 0 8153 4105 5 a b c Helleday T Lo J van Gent DC Engelward BP July 2007 DNA double strand break repair from mechanistic understanding to cancer treatment DNA Repair 6 7 923 35 doi 10 1016 j dnarep 2007 02 006 PMID 17363343 a b Andersen SL Sekelsky J December 2010 Meiotic versus mitotic recombination two different routes for double strand break repair the different functions of meiotic versus mitotic DSB repair are reflected in different pathway usage and different outcomes BioEssays 32 12 1058 66 doi 10 1002 bies 201000087 PMC 3090628 PMID 20967781 Allers T Lichten M July 2001 Differential timing and control of noncrossover and crossover recombination during meiosis Cell 106 1 47 57 doi 10 1016 s0092 8674 01 00416 0 PMID 11461701 S2CID 1878863 Haber lab Single strand annealing Brandeis University Retrieved 3 July 2010 a b c Lyndaker AM Alani E March 2009 A tale of tails insights into the coordination of 3 end processing during homologous recombination BioEssays 31 3 315 21 doi 10 1002 bies 200800195 PMC 2958051 PMID 19260026 Mimitou EP Symington LS September 2009 DNA end resection many nucleases make light work DNA Repair 8 9 983 95 doi 10 1016 j dnarep 2009 04 017 PMC 2760233 PMID 19473888 Paques F Haber JE June 1999 Multiple pathways of recombination induced by double strand breaks in Saccharomyces cerevisiae Microbiology and Molecular Biology Reviews 63 2 349 404 doi 10 1128 MMBR 63 2 349 404 1999 PMC 98970 PMID 10357855 a b McEachern MJ Haber JE 2006 Break induced replication and recombinational telomere elongation in yeast Annual Review of Biochemistry 75 111 35 doi 10 1146 annurev biochem 74 082803 133234 PMID 16756487 Morrish TA Greider CW January 2009 Haber JE ed Short telomeres initiate telomere recombination in primary and tumor cells PLOS Genetics 5 1 e1000357 doi 10 1371 journal pgen 1000357 PMC 2627939 PMID 19180191 nbsp Muntoni A Reddel RR October 2005 The first molecular details of ALT in human tumor cells Human Molecular Genetics 14 Spec No 2 Review Issue 2 R191 6 doi 10 1093 hmg ddi266 PMID 16244317 PDB 3cmt Chen Z Yang H Pavletich NP May 2008 Mechanism of homologous recombination from the RecA ssDNA dsDNA structures Nature 453 7194 489 4 Bibcode 2008Natur 453 489C doi 10 1038 nature06971 PMID 18497818 S2CID 4416531 Kowalczykowski SC Dixon DA Eggleston AK Lauder SD Rehrauer WM September 1994 Biochemistry of homologous recombination in Escherichia coli Microbiological Reviews 58 3 401 65 doi 10 1128 MMBR 58 3 401 465 1994 PMC 372975 PMID 7968921 Rocha EP Cornet E Michel B August 2005 Comparative and evolutionary analysis of the bacterial homologous recombination systems PLOS Genetics 1 2 e15 doi 10 1371 journal pgen 0010015 PMC 1193525 PMID 16132081 nbsp a b Amundsen SK Taylor AF Reddy M Smith GR December 2007 Intersubunit signaling in RecBCD enzyme a complex protein machine regulated by Chi hot spots Genes amp Development 21 24 3296 307 doi 10 1101 gad 1605807 PMC 2113030 PMID 18079176 Singleton MR Dillingham MS Gaudier M Kowalczykowski SC Wigley DB November 2004 Crystal structure of RecBCD enzyme reveals a machine for processing DNA breaks PDF Nature 432 7014 187 93 Bibcode 2004Natur 432 187S doi 10 1038 nature02988 PMID 15538360 S2CID 2916995 Archived from the original PDF on 2004 05 25 Cromie GA August 2009 Phylogenetic ubiquity and shuffling of the bacterial RecBCD and AddAB recombination complexes Journal of Bacteriology 191 16 5076 84 doi 10 1128 JB 00254 09 PMC 2725590 PMID 19542287 a b Smith GR June 2012 How RecBCD enzyme and Chi promote DNA break repair and recombination a molecular biologist s view Microbiology and Molecular Biology Reviews 76 2 217 28 doi 10 1128 MMBR 05026 11 PMC 3372252 PMID 22688812 a b c d Dillingham MS Kowalczykowski SC December 2008 RecBCD enzyme and the repair of double stranded DNA breaks Microbiology and Molecular Biology Reviews 72 4 642 71 Table of Contents doi 10 1128 MMBR 00020 08 PMC 2593567 PMID 19052323 Michel B Boubakri H Baharoglu Z LeMasson M Lestini R July 2007 Recombination proteins and rescue of arrested replication forks DNA Repair 6 7 967 80 doi 10 1016 j dnarep 2007 02 016 PMID 17395553 Spies M Bianco PR Dillingham MS Handa N Baskin RJ Kowalczykowski SC September 2003 A molecular throttle the recombination hotspot chi controls DNA translocation by the RecBCD helicase Cell 114 5 647 54 doi 10 1016 S0092 8674 03 00681 0 PMID 13678587 S2CID 16662983 Taylor AF Smith GR June 2003 RecBCD enzyme is a DNA helicase with fast and slow motors of opposite polarity Nature 423 6942 889 93 Bibcode 2003Natur 423 889T doi 10 1038 nature01674 PMID 12815437 S2CID 4302346 Spies M Amitani I Baskin RJ Kowalczykowski SC November 2007 RecBCD enzyme switches lead motor subunits in response to chi recognition Cell 131 4 694 705 doi 10 1016 j cell 2007 09 023 PMC 2151923 PMID 18022364 Savir Y Tlusty T November 2010 RecA mediated homology search as a nearly optimal signal detection system PDF Molecular Cell 40 3 388 96 arXiv 1011 4382 Bibcode 2010arXiv1011 4382S doi 10 1016 j molcel 2010 10 020 PMID 21070965 S2CID 1682936 Archived from the original PDF on 2012 10 07 Retrieved 2011 08 31 Rambo RP Williams GJ Tainer JA November 2010 Achieving fidelity in homologous recombination despite extreme complexity informed decisions by molecular profiling PDF Molecular Cell 40 3 347 8 doi 10 1016 j molcel 2010 10 032 PMC 3003302 PMID 21070960 Archived from the original PDF on 2012 10 07 Retrieved 2011 08 31 De Vlaminck I van Loenhout MT Zweifel L den Blanken J Hooning K Hage S et al June 2012 Mechanism of homology recognition in DNA recombination from dual molecule experiments Molecular Cell 46 5 616 24 doi 10 1016 j molcel 2012 03 029 PMID 22560720 Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2008 Molecular Biology of the Cell 5th ed Garland Science p 307 ISBN 978 0 8153 4105 5 Morimatsu K Kowalczykowski SC May 2003 RecFOR proteins load RecA protein onto gapped DNA to accelerate DNA strand exchange a universal step of recombinational repair Molecular Cell 11 5 1337 47 doi 10 1016 S1097 2765 03 00188 6 PMID 12769856 Hiom K July 2009 DNA repair common approaches to fixing double strand breaks Current Biology 19 13 R523 5 doi 10 1016 j cub 2009 06 009 PMID 19602417 S2CID 2221866 Handa N Morimatsu K Lovett ST Kowalczykowski SC May 2009 Reconstitution of initial steps of dsDNA break repair by the RecF pathway of E coli Genes amp Development 23 10 1234 45 doi 10 1101 gad 1780709 PMC 2685532 PMID 19451222 a b West SC June 2003 Molecular views of recombination proteins and their control Nature Reviews Molecular Cell Biology 4 6 435 45 doi 10 1038 nrm1127 PMID 12778123 S2CID 28474965 a b c d Watson JD Baker TA Bell SP Gann A Levine M Losick R 2003 Molecular Biology of the Gene 5th ed Pearson Benjamin Cummings pp 259 291 ISBN 978 0 8053 4635 0 Gumbiner Russo LM Rosenberg SM 28 November 2007 Sandler S ed Physical analyses of E coli heteroduplex recombination products in vivo on the prevalence of 5 and 3 patches PLOS ONE 2 11 e1242 Bibcode 2007PLoSO 2 1242G doi 10 1371 journal pone 0001242 PMC 2082072 PMID 18043749 nbsp Thomas CM Nielsen KM September 2005 Mechanisms of and barriers to horizontal gene transfer between bacteria PDF Nature Reviews Microbiology 3 9 711 21 doi 10 1038 nrmicro1234 PMID 16138099 S2CID 1231127 Archived from the original PDF on 2010 06 01 Vulic M Dionisio F Taddei F Radman M September 1997 Molecular keys to speciation DNA polymorphism and the control of genetic exchange in enterobacteria Proceedings of the National Academy of Sciences of the United States of America 94 18 9763 7 Bibcode 1997PNAS 94 9763V doi 10 1073 pnas 94 18 9763 PMC 23264 PMID 9275198 Majewski J Cohan FM January 1998 The effect of mismatch repair and heteroduplex formation on sexual isolation in Bacillus Genetics 148 1 13 8 doi 10 1093 genetics 148 1 13 PMC 1459767 PMID 9475717 Majewski J Zawadzki P Pickerill P Cohan FM Dowson CG February 2000 Barriers to genetic exchange between bacterial species Streptococcus pneumoniae transformation Journal of Bacteriology 182 4 1016 23 doi 10 1128 JB 182 4 1016 1023 2000 PMC 94378 PMID 10648528 Chen I Dubnau D March 2004 DNA uptake during bacterial transformation Nature Reviews Microbiology 2 3 241 9 doi 10 1038 nrmicro844 PMID 15083159 S2CID 205499369 Claverys JP Martin B Polard P May 2009 The genetic transformation machinery composition localization and mechanism FEMS Microbiology Reviews 33 3 643 56 doi 10 1111 j 1574 6976 2009 00164 x PMID 19228200 Kidane D Graumann PL July 2005 Intracellular protein and DNA dynamics in competent Bacillus subtilis cells Cell 122 1 73 84 doi 10 1016 j cell 2005 04 036 PMID 16009134 S2CID 17272331 Fleischmann Jr WR 1996 43 Medical Microbiology 4th ed University of Texas Medical Branch at Galveston ISBN 978 0 9631172 1 2 Boni MF de Jong MD van Doorn HR Holmes EC 3 May 2010 Martin DP ed Guidelines for identifying homologous recombination events in influenza A virus PLOS ONE 5 5 e10434 Bibcode 2010PLoSO 510434B doi 10 1371 journal pone 0010434 PMC 2862710 PMID 20454662 nbsp a b Nagy PD Bujarski JJ January 1996 Homologous RNA recombination in brome mosaic virus AU rich sequences decrease the accuracy of crossovers Journal of Virology 70 1 415 26 doi 10 1128 JVI 70 1 415 426 1996 PMC 189831 PMID 8523555 Chetverin AB October 1999 The puzzle of RNA recombination FEBS Letters 460 1 1 5 doi 10 1016 S0014 5793 99 01282 X PMC 7163957 PMID 10571050 a b Roossinck MJ September 1997 Mechanisms of plant virus evolution Annual Review of Phytopathology 35 191 209 doi 10 1146 annurev phyto 35 1 191 PMID 15012521 Arbuckle JH Medveczky PG August 2011 The molecular biology of human herpesvirus 6 latency and telomere integration Microbes and Infection Institut Pasteur 13 8 9 731 41 doi 10 1016 j micinf 2011 03 006 PMC 3130849 PMID 21458587 Bernstein C March 1981 Deoxyribonucleic acid repair in bacteriophage Microbiological Reviews 45 1 72 98 doi 10 1128 MMBR 45 1 72 98 1981 PMC 281499 PMID 6261109 Bernstein C Bernstein H 2001 DNA repair in bacteriophage In Nickoloff JA Hoekstra MF Eds DNA Damage and Repair Vol 3 Advances from Phage to Humans Humana Press Totowa NJ pp 1 19 ISBN 978 0896038035 Story RM Bishop DK Kleckner N Steitz TA March 1993 Structural relationship of bacterial RecA proteins to recombination proteins from bacteriophage T4 and yeast Science 259 5103 1892 6 Bibcode 1993Sci 259 1892S doi 10 1126 science 8456313 PMID 8456313 Michod RE Bernstein H Nedelcu AM May 2008 Adaptive value of sex in microbial pathogens Infection Genetics and Evolution 8 3 267 85 doi 10 1016 j meegid 2008 01 002 PMID 18295550 http www hummingbirds arizona edu Faculty Michod Downloads IGE 20review 20sex pdf a b Su Shuo Wong Gary Shi Weifeng Liu Jun Lai Alexander C K Zhou Jiyong Liu Wenjun Bi Yuhai Gao George F 2016 Epidemiology Genetic Recombination and Pathogenesis of Coronaviruses Trends in Microbiology 24 6 490 502 doi 10 1016 j tim 2016 03 003 PMC 7125511 PMID 27012512 Barr J N Fearns R 2010 How RNA viruses maintain their genome integrity Journal of General Virology 91 6 1373 1387 doi 10 1099 vir 0 020818 0 PMID 20335491 a b Li Xiaojun Giorgi Elena E Marichannegowda Manukumar Honnayakanahalli Foley Brian Xiao Chuan Kong Xiang Peng Chen Yue Gnanakaran S Korber Bette Gao Feng 2020 Emergence of SARS CoV 2 through recombination and strong purifying selection Science Advances 6 27 Bibcode 2020SciA 6 9153L doi 10 1126 sciadv abb9153 PMC 7458444 PMID 32937441 Rehman Saif ur Shafique Laiba Ihsan Awais Liu Qingyou 2020 Evolutionary Trajectory for the Emergence of Novel Coronavirus SARS CoV 2 Pathogens 9 3 240 doi 10 3390 pathogens9030240 PMC 7157669 PMID 32210130 Yeh TY Contreras GP July 2020 Emerging viral mutants in Australia suggest RNA recombination event in the SARS CoV 2 genome The Medical Journal of Australia 213 1 44 44 e1 doi 10 5694 mja2 50657 PMC 7300921 PMID 32506536 Yeh TY Contreras GP 1 July 2021 Viral transmission and evolution dynamics of SARS CoV 2 in shipboard quarantine Bull World Health Organ 99 7 486 495 doi 10 2471 BLT 20 255752 PMC 8243027 PMID 34248221 Lamb NE Yu K Shaffer J Feingold E Sherman SL January 2005 Association between maternal age and meiotic recombination for trisomy 21 American Journal of Human Genetics 76 1 91 9 doi 10 1086 427266 PMC 1196437 PMID 15551222 Cold Spring Harbor Laboratory 2007 Human RecQ Helicases Homologous Recombination And Genomic Instability ScienceDaily Retrieved 3 July 2010 Modesti M Kanaar R 2001 Homologous recombination from model organisms to human disease Genome Biology 2 5 REVIEWS1014 doi 10 1186 gb 2001 2 5 reviews1014 PMC 138934 PMID 11387040 Luo G Santoro IM McDaniel LD Nishijima I Mills M Youssoufian H Vogel H Schultz RA Bradley A December 2000 Cancer predisposition caused by elevated mitotic recombination in Bloom mice Nature Genetics 26 4 424 9 doi 10 1038 82548 PMID 11101838 S2CID 21218975 a b Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2007 Molecular Biology of the Cell 5th ed Garland Science ISBN 978 0 8153 4110 9 a b c Powell SN Kachnic LA September 2003 Roles of BRCA1 and BRCA2 in homologous recombination DNA replication fidelity and the cellular response to ionizing radiation Oncogene 22 37 5784 91 doi 10 1038 sj onc 1206678 PMID 12947386 Use of homologous recombination deficiency HRD score to enrich for niraparib sensitive high grade ovarian tumors Archived from the original on 2017 04 30 Retrieved 2016 12 30 a b c d e f g h Lin Z Kong H Nei M Ma H July 2006 Origins and evolution of the recA RAD51 gene family evidence for ancient gene duplication and endosymbiotic gene transfer Proceedings of the National Academy of Sciences of the United States of America 103 27 10328 33 Bibcode 2006PNAS 10310328L doi 10 1073 pnas 0604232103 PMC 1502457 PMID 16798872 a b Haseltine CA Kowalczykowski SC May 2009 An archaeal Rad54 protein remodels DNA and stimulates DNA strand exchange by RadA Nucleic Acids Research 37 8 2757 70 doi 10 1093 nar gkp068 PMC 2677860 PMID 19282450 Rolfsmeier ML Haseltine CA March 2010 The single stranded DNA binding protein of Sulfolobus solfataricus acts in the presynaptic step of homologous recombination Journal of Molecular Biology 397 1 31 45 doi 10 1016 j jmb 2010 01 004 PMID 20080104 Huang Q Liu L Liu J Ni J She Q Shen Y 2015 Efficient 5 3 DNA end resection by HerA and NurA is essential for cell viability in the crenarchaeon Sulfolobus islandicus BMC Molecular Biology 16 2 doi 10 1186 s12867 015 0030 z PMC 4351679 PMID 25880130 nbsp Jain SK Cox MM Inman RB August 1994 On the role of ATP hydrolysis in RecA protein mediated DNA strand exchange III Unidirectional branch migration and extensive hybrid DNA formation The Journal of Biological Chemistry 269 32 20653 61 doi 10 1016 S0021 9258 17 32043 4 PMID 8051165 Ramesh MA Malik SB Logsdon JM January 2005 A phylogenomic inventory of meiotic genes evidence for sex in Giardia and an early eukaryotic origin of meiosis Current Biology 15 2 185 91 doi 10 1016 j cub 2005 01 003 PMID 15668177 S2CID 17013247 a b Malik SB Ramesh MA Hulstrand AM Logsdon JM December 2007 Protist homologs of the meiotic Spo11 gene and topoisomerase VI reveal an evolutionary history of gene duplication and lineage specific loss Molecular Biology and Evolution 24 12 2827 41 doi 10 1093 molbev msm217 PMID 17921483 Lodish H Berk A Zipursky SL Matsudaira P Baltimore D Darnell J 2000 Chapter 8 5 Gene Replacement and Transgenic Animals DNA Is Transferred into Eukaryotic Cells in Various Ways Molecular Cell Biology 4th ed W H Freeman and Company ISBN 978 0 7167 3136 8 The Nobel Prize in Physiology or Medicine 2007 The Nobel Foundation Retrieved December 15 2008 Drummond DA Silberg JJ Meyer MM Wilke CO Arnold FH April 2005 On the conservative nature of intragenic recombination Proceedings of the National Academy of Sciences of the United States of America 102 15 5380 5 Bibcode 2005PNAS 102 5380D doi 10 1073 pnas 0500729102 PMC 556249 PMID 15809422 Bloom JD Silberg JJ Wilke CO Drummond DA Adami C Arnold FH January 2005 Thermodynamic prediction of protein neutrality Proceedings of the National Academy of Sciences of the United States of America 102 3 606 11 arXiv q bio 0409013 Bibcode 2005PNAS 102 606B doi 10 1073 pnas 0406744102 PMC 545518 PMID 15644440 a b Carbone MN Arnold FH August 2007 Engineering by homologous recombination exploring sequence and function within a conserved fold Current Opinion in Structural Biology 17 4 454 9 doi 10 1016 j sbi 2007 08 005 PMID 17884462 Otey CR Landwehr M Endelman JB Hiraga K Bloom JD Arnold FH May 2006 Structure guided recombination creates an artificial family of cytochromes P450 PLOS Biology 4 5 e112 doi 10 1371 journal pbio 0040112 PMC 1431580 PMID 16594730 nbsp Socolich M Lockless SW Russ WP Lee H Gardner KH Ranganathan R September 2005 Evolutionary information for specifying a protein fold Nature 437 7058 512 8 Bibcode 2005Natur 437 512S doi 10 1038 nature03991 PMID 16177782 S2CID 4363255 Thulasiram HV Erickson HK Poulter CD April 2007 Chimeras of two isoprenoid synthases catalyze all four coupling reactions in isoprenoid biosynthesis Science 316 5821 73 6 Bibcode 2007Sci 316 73T doi 10 1126 science 1137786 PMID 17412950 S2CID 43516273 Landwehr M Carbone M Otey CR Li Y Arnold FH March 2007 Diversification of catalytic function in a synthetic family of chimeric cytochrome p450s Chemistry amp Biology 14 3 269 78 doi 10 1016 j chembiol 2007 01 009 PMC 1991292 PMID 17379142 Choudhury Ananya Zhao Helen Jalali Farid Rashid Shahnaz AL Ran Jane Supiot Stephane Kiltie Anne E Bristow Robert G 1 January 2009 Targeting homologous recombination using imatinib results in enhanced tumor cell chemosensitivity and radiosensitivity PDF Molecular Cancer Therapeutics 8 1 203 213 doi 10 1158 1535 7163 MCT 08 0959 ISSN 1535 7163 PMID 19139130 S2CID 6493434 Siddiqui Arafat Tumiati Manuela Joko Alia Sandholm Jouko Roering Pia Aakko Sofia Vainionpaa Reetta Kaipio Katja Huhtinen Kaisa Kauppi Liisa Tuomela Johanna Hietanen Sakari 2021 Targeting DNA Homologous Repair Proficiency With Concomitant Topoisomerase II and c Abl Inhibition Frontiers in Oncology 11 3666 doi 10 3389 fonc 2021 733700 ISSN 2234 943X PMC 8488401 PMID 34616682 a b c Iglehart JD Silver DP July 2009 Synthetic lethality a new direction in cancer drug development The New England Journal of Medicine 361 2 189 91 doi 10 1056 NEJMe0903044 PMID 19553640 Fong PC Boss DS Yap TA Tutt A Wu P Mergui Roelvink M Mortimer P Swaisland H Lau A O Connor MJ Ashworth A Carmichael J Kaye SB Schellens JH de Bono JS July 2009 Inhibition of poly ADP ribose polymerase in tumors from BRCA mutation carriers The New England Journal of Medicine 361 2 123 34 doi 10 1056 NEJMoa0900212 PMID 19553641 Edwards SL Brough R Lord CJ Natrajan R Vatcheva R Levine DA Boyd J Reis Filho JS Ashworth A February 2008 Resistance to therapy caused by intragenic deletion in BRCA2 Nature 451 7182 1111 5 Bibcode 2008Natur 451 1111E doi 10 1038 nature06548 PMID 18264088 S2CID 205212044 External links edit nbsp Wikimedia Commons has media related to Homologous recombination Animations homologous recombination Animations showing several models of homologous recombination Homologous recombination Tempy amp Trun Animation of the bacterial RecBCD pathway of homologous recombination Retrieved from https en wikipedia org w index php title Homologous recombination amp oldid 1204298015, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.