fbpx
Wikipedia

Type II topoisomerase

Type II topoisomerases are topoisomerases that cut both strands of the DNA helix simultaneously in order to manage DNA tangles and supercoils. They use the hydrolysis of ATP, unlike Type I topoisomerase. In this process, these enzymes change the linking number of circular DNA by ±2. Topoisomerases are ubiquitous enzymes, found in all living organisms.[1]

DNA Topoisomerase II (ATP-hydrolyzing)
Structure of the 42 KDa fragment of the N-terminal ATPase and transducer domains of DNA gyrase homologous to all other type IIA topoisomerases.
Identifiers
EC no.5.6.2.2
Databases
IntEnzIntEnz view
BRENDABRENDA entry
ExPASyNiceZyme view
KEGGKEGG entry
MetaCycmetabolic pathway
PRIAMprofile
PDB structuresRCSB PDB PDBe PDBsum
Search
PMCarticles
PubMedarticles
NCBIproteins

In animals, topoisomerase II is a chemotherapy target. In prokaryotes, gyrase is an antibacterial target.[2] Indeed, these enzymes are of interest for a wide range of effects.

Function edit

Type II topoisomerases increase or decrease the linking number of a DNA loop by 2 units, and it promotes chromosome disentanglement. For example, DNA gyrase, a type II topoisomerase observed in E. coli and most other prokaryotes, introduces negative supercoils and decreases the linking number by 2. Gyrase is also able to remove knots from the bacterial chromosome. Along with gyrase, most prokaryotes also contain a second type IIA topoisomerase, termed topoisomerase IV. Gyrase and topoisomerase IV differ by their C-terminal domains, which is believed to dictate substrate specificity and functionality for these two enzymes. Footprinting indicates that gyrase, which forms a 140-base-pair footprint and wraps DNA, introduces negative supercoils, while topoisomerase IV, which forms a 28-base-pair footprint, does not wrap DNA.

Eukaryotic type II topoisomerase cannot introduce supercoils; it can only relax them.

The roles of type IIB topoisomerases are less understood. Unlike type IIA topoisomerases, type IIB topoisomerases cannot simplify DNA topology (see below), but they share several structural features with type IIA topoisomerases.

Topology simplification edit

Type IIA topoisomerases are essential in the separation of entangled daughter strands during replication. This function is believed to be performed by topoisomerase II in eukaryotes and by topoisomerase IV in prokaryotes. Failure to separate these strands leads to cell death. Type IIA topoisomerases have the special ability to relax DNA to a state below that of thermodynamic equilibrium, a feature unlike type IA, IB, and IIB topoisomerases. This ability, known as topology simplification, was first identified by Rybenkov et al.[3] The hydrolysis of ATP drives this simplification, but a clear molecular mechanism for this simplification is still lacking. Several models to explain this phenomenon have been proposed, including two models that rely on the ability of type IIA topoisomerases to recognize bent DNA duplexes.[4] Biochemistry, electron microscopy, and recent structures of topoisomerase II bound to DNA reveal that type IIA topoisomerases bind at the apices of DNA, supporting this model.

Classification edit

There are two subclasses of type II topoisomerases, type IIA and IIB.

  • Type IIA topoisomerases include the enzymes DNA gyrase, eukaryotic topoisomerase II (topo II), and bacterial topoisomerase IV (topo IV). These enzymes span all domains of life and are essential for function.[5]
  • Type IIB topoisomerases are structurally and biochemically distinct, and comprise a single family member, topoisomerase VI (topo VI). Type IIB topoisomerases are found in archaea and some higher plants.

Some organisms including humans have two isoforms of topoisomerase II: alpha and beta. In cancers, the topoisomerase IIα is highly expressed in proliferating cells. In certain cancers, such as peripheral nerve sheath tumors, high expression of its encoded protein is also associated to poor patient survival.

The two classes of topoisomerases possess a similar strand passage mechanism and domain structure (see below), however they also have several important differences. Type IIA topoisomerases form double-stranded breaks with four-base pair overhangs, while type IIB topoisomerases form double-stranded breaks with two base overhangs.[6] In addition, type IIA topoisomerases are able to simplify DNA topology,[3] while type IIB topoisomerases do not.[7]

Structure edit

Type IIA edit

 
Schematic structure of gyrase, oriented upside-down compared to the other examples in this article.
 
Structure of yeast topoisomerase II bound to a doubly nicked 34-mer duplex DNA (PDB: 2RGR​). The Toprim fold is colored cyan; the DNA is colored orange; the HTH is colored magenta; and the C-gate is colored purple. Notice that the DNA is bent by ~160 degrees through an invariant isoleucine (Ile833 in yeast).

Type IIA topoisomerases consist of several key motifs:

  • an N-terminal GHKL ATPase domain (for gyrase, Hsp, kinase and MutL),
  • a Toprim domain (a Rossmann fold subclass), which exists in both type II topoisomerases, type IA topoisomerases, and bacterial primase (DnaG),
  • a central DNA-binding core (which structurally forms a heart-shaped structure), and
  • a variable C-terminal domain.

Eukaryotic type II topoisomerases are homodimers (A2), while prokaryotic type II topoisomerases are heterotetramers (A2B2). Prokaryotes have the ATPase domain and the Toprim fold on one polypeptide (Pfam PF00204), while the DNA cleavage core and the CTD lies on a second polypeptide (Pfam PF00521). For gyrase, the first polypeptide is called GyrB and the second polypeptide is called GyrA. For topo IV, the first polypeptide is called ParE and the second polypeptide is called ParC. Both Pfam signatures are found in the single-chain eukayotic topoisomerase.

The structures of the N-terminal ATPase domain of gyrase[8] and yeast topoisomerase II[9] have been solved in complex with AMPPNP (an ATP analogue), showing that two ATPase domains dimerize to form a closed conformation. For gyrase, the structure has a substantial hole in the middle, which is presumed to accommodate the T-segment.

Linking the ATPase domain to the Toprim fold is a helical element known as the transducer domain. This domain is thought to communicate the nucleotide state of the ATPase domain to the rest of the protein. Modifications to this domain affect topoisomerase activity, and structural work done by the Verdine group shows that the ATP state affects the orientation of the transducer domain.[10]

The central core of the protein contains a Toprim fold and a DNA-binding core that contains a winged helix domain (WHD), often referred to as a CAP domain, since it was first identified to resemble the WHD of catabolite activator protein. The catalytic tyrosine lies on this WHD. The Toprim fold is a Rossmann fold that contains three invariant acidic residues that coordinate magnesium ions involved in DNA cleavage and DNA religation.[11] The structure of the Toprim fold and DNA-binding core of yeast topoisomerase II was first solved by Berger and Wang,[12] and the first gyrase DNA-binding core was solved by Morais Cabral et al.[13] The structure solved by Berger revealed important insights into the function of the enzyme. The DNA-binding core consists of the WHD, which leads to a tower domain. A coiled-coil region leads to a C-terminal domain that forms the main dimer interface for this crystal state (often termed the C-gate). While the original topoisomerase II structure shows a situation where the WHDs are separated by a large distance, the structure of gyrase shows a closed conformation, where the WHD close.

The topoisomerase II core was later solved in new conformations, including one by Fass et al.[14] and one by Dong et al.[15] The Fass structure shows that the Toprim domain is flexible and that this flexibility can allow the Toprim domain to coordinate with the WHD to form a competent cleavage complex. This was eventually substantiated by the Dong et al. structure that was solved in the presence of DNA. This last structure showed that the Toprim domain and the WHD formed a cleavage complex very similar to that of the type IA topoisomerases and indicated how DNA-binding and cleavage could be uncoupled, and the structure showed that DNA was bent by ~150 degrees through an invariant isoleucine (in topoisomerase II it is I833 and in gyrase it is I172). This mechanism of bending resembles closely that of integration host factor (IHF) and HU, two architectural proteins in bacteria. In addition, while the previous structures of the DNA-binding core had the C-gate closed, this structure captured the gate open, a key step in the two-gate mechanism (see below).

More recently, several structures of the DNA-bound structure have been solved in an attempt to understand both the chemical mechanism for DNA cleavage and the structural basis for inhibition of topoisomerase by antibacterial poisons. The first complete architecture of the E. coli DNA gyrase has been solved by cryo-electron microscopy at near atomic resolution.[16] The nucleoprotein complex was captured with a long DNA duplex and gepotidacin, a novel bacterial topoisomerase inhibitor.

The C-terminal region of the prokaryotic topoisomerases has been solved for multiple species. The first structure of a C-terminal domain of gyrase was solved by Corbett et al.[17] and the C-terminal domain of topoisomerase IV was solved by Corbett et al.[7] The structures formed a novel beta barrel, which bends DNA by wrapping the nucleic acid around itself. The bending of DNA by gyrase has been proposed as a key mechanism in the ability of gyrase to introduce negative supercoils into the DNA. This is consistent with footprinting data that shows that gyrase has a 140-base-pair footprint. Both gyrase and topoisomerase IV CTDs bend DNA, but only gyrase introduces negative supercoils.

Unlike the function of the C-terminal domain of prokaryotic topoisomerases, the function of the C-terminal region of eukaryotic topoisomerase II is still not clear. Studies have suggested that this region is regulated by phosphorylation and this modulates topoisomerase activity, however more research needs to be done to investigate this.

Type IIB edit

 
Structure of topo VI (PDB: 2Q2E​) in an orientation similar to the yeast example. Chains are colored differently. The Toprim domain lies on the top, and the ATPase domain lies on the bottom; each forms a DNA gate.

The organization of type IIB topoisomerases are similar to that of type IIAs, except that all type IIBs have two genes and form heterotetramers. One gene, termed topo VI-B (since it resembles gyrB), contains the ATPase domain, a transducer domain (Pfam PF09239), and a C-terminal Ig-fold-like H2TH domain (Pfam PF18000). The second gene, termed topo VI-A (Pfam PF04406), contains the WHD and the Toprim domain.

The ATPase domain of topo VI B was solved in multiple nucleotide states.[18] It closely resembles that of the GHKL domain of topo II and MutL and shows that the nucleotide state (ADP versus ATP) effects the orientation of the transducer domain ( and 1MX0).

The structure of topo VI-A was solved by Bergerat et al.[19] showing that the HTH and Toprim fold had a novel conformation compared with that of topo IIA.

A recent structure of the topo VI A/B complex was solved, showing an open and closed conformation, two states that are predicted in the two-gate mechanism (see below). These structures, of which one is an X-ray crystal structure and the other is a Small-Angle X-ray Scattering (SAXS) reconstruction, show that the ATPase domain can be either open or closed.[20]

Mechanism of action edit

Strand passage edit

Type IIA topoisomerase operates through a "two-gate" mechanism (though this is a historical notation), a mechanism supported by biochemistry[21] as well as by structural work.[22]

A strand of DNA, called the gate, or G-segment, is bound by a central DNA-binding gate (DNA-gate). A second strand of DNA, called the transport, or T-segment, is captured by the dimerization of the N-terminal ATPase domain (the ATPase-gate) when two molecules of ATP bind. Hydrolysis of ATP and release of an inorganic phosphate leads to the cleavage of the G-segment, as the catalytic tyrosines form a covalent phosphotyrosine bond with the 5' end of the DNA. This creates a four-base overhang and a double-stranded break in the G-segment. As the DNA-binding gate separates, the T-segment is transferred through the G-segment. The G-segment is sealed, leading to the C-terminal gate (or C-gate) to open, allowing for the release of the T-segment. Release of product ADP leads to a reset of the system, and allows a second T-segment to be captured.

Type IIB topoisomerases operate through a similar fashion, except that the protein forms a two-base overhang in the G-segment and the C-terminal gate is completely missing.

DNA cleavage edit

In the strand passage mechanism, the cleavage of DNA is key to allow the T-segment to transfer through the G-segment. The mechanism of DNA cleavage by type IIA topoisomerases has recently been the focus of many biochemical and structural biology studies.

Catenation edit

Catenation is the process by which two circular DNA strands are linked together like chain links. This occurs after DNA replication, where two single strands are catenated and can still replicate but cannot separate into the two daughter cells. As type II topoisomerses break a double strand, they can fix this state (type I topoisomerases could do this only if there were already a single-strand nick), and the correct chromosome number can remain in daughter cells. Linear DNA in eukaryotes is so long they can be thought of as being without ends; type II topoisomerases are needed for the same reason.

Inhibition edit

Small molecules that target type II topoisomerase are divided into two classes: inhibitors and poisons. Due to their frequent presence in proliferating eukaryotic cells, inhibitors of type II topoisomerases have been extensively studied and used as anti-cancer medications.[23]

  • Inhibitors of type II topoisomerase include HU-331, ICRF-187, ICRF-193, and mitindomide. These molecules work by inhibiting the ATPase activity by acting as noncompetitive inhibitors of ATP. This has been shown through structural studies[9] and biochemical studies performed by the Lindsley group.
  • Poisons of type II topoisomerases include doxorubicin, etoposide, novobiocin, quinolones (including ciprofloxacin), and teniposide. These small molecules target the DNA-protein complex. Some of these molecules lead to increased cleavage, whereas others, such as etoposide, inhibit religation.

The experimental antitumor drug m-AMSA (4'-(9'-acridinylamino)methanesulfon-m-anisidide) also inhibits type 2 topoisomerase.[24]

Topoisomerase poisons have been extensively used as both anticancer and antibacterial therapies. While antibacterial compounds such as ciprofloxacin target bacterial gyrase, they fail to inhibit eukaryotic type IIA topoisomerases. In addition, drug-resistant bacteria often have a point mutation in gyrase (Serine79Alanine in E. coli) that renders quinolones ineffective.[citation needed] Recent structural studies have led to the discovery of a compound that no longer relies on this residue and, therefore, has efficacy against drug-resistant bacteria.[citation needed]

Bacteriophage T4 gyrase edit

The bacteriophage (phage) T4 gyrase (type II topoismerase) is a multisubunit protein consisting of the products of genes 39, 52 and probably 60.[25][26] It catalyses the relaxation of negatively or positively superhelical DNA and is employed in phage DNA replication during infection of the E. coli bacterial host.[27] The phage gene 52 protein shares homology with the E. coli gyrase gyrA subunit[28] and the phage gene 39 protein shares homology with the gyr B subunit.[29] Since the host E. coli DNA gyrase can partially compensate for the loss of the phage T4 gene products, mutants defective in either genes 39, 52 or 60 do not completely abolish phage DNA replication, but rather delay its initiation.[27] The rate of DNA elongation is not slower than wild-type in such mutant infections.[30] Mutants defective in genes 39, 52 or 60 show increased genetic recombination as well as increased base-substitution and deletion mutation suggesting that the host compensated DNA synthesis is less accurate than that directed by wild-type phage.[31] A mutant defective in gene 39 shows increased sensitivity to inactivation by ultraviolet irradiation during the stage of phage infection after initiation of DNA replication when multiple copies of the phage chromosome are present.[32] Mutants defective in genes 39, 52 and 60 have reduced ability to carry out multiplicity reactivation, a form of recombinational repair that can deal with different types of DNA damage.[33] The gyrase specified by the genome of uninfected E. coli also appears to participate in recombinational repair by providing an initiation point for the reciprocal strand exchange driven by the RecA protein.[34]

References edit

  1. ^ Deweese JE, Osheroff N (February 2009). "The DNA cleavage reaction of topoisomerase II: wolf in sheep's clothing". Nucleic Acids Research. 37 (3): 738–748. doi:10.1093/nar/gkn937. PMC 2647315. PMID 19042970.
  2. ^ Reece RJ, Maxwell A (1991). "DNA gyrase: structure and function". Critical Reviews in Biochemistry and Molecular Biology. 26 (3–4): 335–375. doi:10.3109/10409239109114072. PMID 1657531.
  3. ^ a b Rybenkov VV, Ullsperger C, Vologodskii AV, Cozzarelli NR (August 1997). "Simplification of DNA topology below equilibrium values by type II topoisomerases". Science. 277 (5326). New York, N.Y.: 690–3. doi:10.1126/science.277.5326.690. PMID 9235892.
  4. ^ Vologodskii AV, Zhang W, Rybenkov VV, Podtelezhnikov AA, Subramanian D, Griffith JD, Cozzarelli NR (March 2001). "Mechanism of topology simplification by type II DNA topoisomerases". Proceedings of the National Academy of Sciences of the United States of America. 98 (6): 3045–9. Bibcode:2001PNAS...98.3045V. doi:10.1073/pnas.061029098. PMC 30604. PMID 11248029.
  5. ^ Reece RJ, Maxwell A (January 1991). "DNA gyrase: structure and function". Critical Reviews in Biochemistry and Molecular Biology. 26 (3–4): 335–75. doi:10.3109/10409239109114072. PMID 1657531.
  6. ^ Buhler C, Lebbink JH, Bocs C, Ladenstein R, Forterre P (October 2001). "DNA topoisomerase VI generates ATP-dependent double-strand breaks with two-nucleotide overhangs". The Journal of Biological Chemistry. 276 (40): 37215–22. doi:10.1074/jbc.M101823200. PMID 11485995. S2CID 24354635.
  7. ^ a b PDB: 1zvt​; Corbett KD, Schoeffler AJ, Thomsen ND, Berger JM (August 2005). "The structural basis for substrate specificity in DNA topoisomerase IV". Journal of Molecular Biology. 351 (3): 545–61. doi:10.1016/j.jmb.2005.06.029. PMID 16023670.
  8. ^ Wigley DB, Davies GJ, Dodson EJ, Maxwell A, Dodson G (June 1991). "Crystal structure of an N-terminal fragment of the DNA gyrase B protein". Nature. 351 (6328): 624–9. Bibcode:1991Natur.351..624W. doi:10.1038/351624a0. PMID 1646964. S2CID 4373125.
  9. ^ a b PDB: 1PVG​; Classen S, Olland S, Berger JM (September 2003). "Structure of the topoisomerase II ATPase region and its mechanism of inhibition by the chemotherapeutic agent ICRF-187". Proceedings of the National Academy of Sciences of the United States of America. 100 (19): 10629–34. Bibcode:2003PNAS..10010629C. doi:10.1073/pnas.1832879100. PMC 196855. PMID 12963818.
  10. ^ Wei H, Ruthenburg AJ, Bechis SK, Verdine GL (November 2005). "Nucleotide-dependent domain movement in the ATPase domain of a human type IIA DNA topoisomerase" (PDF). The Journal of Biological Chemistry. 280 (44): 37041–7. doi:10.1074/jbc.M506520200. PMID 16100112. S2CID 35186716.
  11. ^ Aravind L, Leipe DD, Koonin EV (September 1998). "Toprim--a conserved catalytic domain in type IA and II topoisomerases, DnaG-type primases, OLD family nucleases and RecR proteins". Nucleic Acids Research. 26 (18): 4205–13. doi:10.1093/nar/26.18.4205. PMC 147817. PMID 9722641.
  12. ^ PDB: 1BGW​; Berger JM, Gamblin SJ, Harrison SC, Wang JC (January 1996). "Structure and mechanism of DNA topoisomerase II". Nature. 379 (6562): 225–32. Bibcode:1996Natur.379..225B. doi:10.1038/379225a0. PMID 8538787. S2CID 4360011.
  13. ^ PDB: 1AB4​; Morais Cabral JH, Jackson AP, Smith CV, Shikotra N, Maxwell A, Liddington RC (August 1997). "Crystal structure of the breakage-reunion domain of DNA gyrase". Nature. 388 (6645): 903–6. Bibcode:1997Natur.388..903M. doi:10.1038/42294. PMID 9278055. S2CID 4320715.
  14. ^ PDB: 1BJT​; Fass D, Bogden CE, Berger JM (April 1999). "Quaternary changes in topoisomerase II may direct orthogonal movement of two DNA strands". Nature Structural Biology. 6 (4): 322–6. doi:10.1038/7556. PMID 10201398. S2CID 947461.
  15. ^ PDB: 2RGR​; Dong KC, Berger JM (December 2007). "Structural basis for gate-DNA recognition and bending by type IIA topoisomerases". Nature. 450 (7173): 1201–5. Bibcode:2007Natur.450.1201D. doi:10.1038/nature06396. PMID 18097402. S2CID 1756317.
  16. ^ Vanden Broeck A, Lotz C, Ortiz J, Lamour V (October 2019). "Cryo-EM structure of the complete E. coli DNA gyrase nucleoprotein complex". Nature Communications. 10 (1): 4935. Bibcode:2019NatCo..10.4935V. doi:10.1038/s41467-019-12914-y. PMC 6821735. PMID 31666516.
  17. ^ PDB: 1SUU​; Corbett KD, Shultzaberger RK, Berger JM (May 2004). "The C-terminal domain of DNA gyrase A adopts a DNA-bending beta-pinwheel fold". Proceedings of the National Academy of Sciences of the United States of America. 101 (19): 7293–8. Bibcode:2004PNAS..101.7293C. doi:10.1073/pnas.0401595101. PMC 409912. PMID 15123801.
  18. ^ PDB: 1MU5​; Corbett KD, Berger JM (January 2003). "Structure of the topoisomerase VI-B subunit: implications for type II topoisomerase mechanism and evolution". The EMBO Journal. 22 (1): 151–63. doi:10.1093/emboj/cdg008. PMC 140052. PMID 12505993.
  19. ^ Bergerat A, de Massy B, Gadelle D, Varoutas PC, Nicolas A, Forterre P (March 1997). "An atypical topoisomerase II from Archaea with implications for meiotic recombination". Nature. 386 (6623): 414–7. Bibcode:1997Natur.386..414B. doi:10.1038/386414a0. PMID 9121560. S2CID 4327493.
  20. ^ PDB: 2Q2E​; Corbett KD, Benedetti P, Berger JM (July 2007). "Holoenzyme assembly and ATP-mediated conformational dynamics of topoisomerase VI". Nature Structural & Molecular Biology. 14 (7): 611–9. doi:10.1038/nsmb1264. PMID 17603498. S2CID 2159631.
  21. ^ Roca J, Wang JC (May 1994). "DNA transport by a type II DNA topoisomerase: evidence in favor of a two-gate mechanism". Cell. 77 (4): 609–16. doi:10.1016/0092-8674(94)90222-4. PMID 8187179. S2CID 19776252.
  22. ^ Berger JM, Wang JC (February 1996). "Recent developments in DNA topoisomerase II structure and mechanism". Current Opinion in Structural Biology. 6 (1): 84–90. doi:10.1016/s0959-440x(96)80099-6. PMID 8696977.
  23. ^ Alberts B (2014-11-18). Molecular biology of the cell (Sixth ed.). New York, NY. ISBN 978-0-8153-4432-2. OCLC 887605755.{{cite book}}: CS1 maint: location missing publisher (link)
  24. ^ Willmore E, de Caux S, Sunter NJ, Tilby MJ, Jackson GH, Austin CA, Durkacz BW (June 2004). "A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia". Blood. 103 (12): 4659–65. doi:10.1182/blood-2003-07-2527. PMID 15010369.
  25. ^ Liu LF, Liu CC, Alberts BM (October 1979). "T4 DNA topoisomerase: a new ATP-dependent enzyme essential for initiation of T4 bacteriophage DNA replication". Nature. 281 (5731): 456–61. Bibcode:1979Natur.281..456L. doi:10.1038/281456a0. PMID 226889. S2CID 4343962.
  26. ^ Stetler GL, King GJ, Huang WM (August 1979). "T4 DNA-delay proteins, required for specific DNA replication, form a complex that has ATP-dependent DNA topoisomerase activity". Proceedings of the National Academy of Sciences of the United States of America. 76 (8): 3737–41. Bibcode:1979PNAS...76.3737S. doi:10.1073/pnas.76.8.3737. PMC 383908. PMID 226976.
  27. ^ a b McCarthy D (January 1979). "Gyrase-dependent initiation of bacteriophage T4 DNA replication: interactions of Escherichia coli gyrase with novobiocin, coumermycin and phage DNA-delay gene products". Journal of Molecular Biology. 127 (3): 265–83. doi:10.1016/0022-2836(79)90329-2. PMID 372540.
  28. ^ Huang WM (September 1986). "The 52-protein subunit of T4 DNA topoisomerase is homologous to the gyrA-protein of gyrase". Nucleic Acids Research. 14 (18): 7379–90. PMC 311757. PMID 3020513.
  29. ^ Huang WM (October 1986). "Nucleotide sequence of a type II DNA topoisomerase gene. Bacteriophage T4 gene 39". Nucleic Acids Research. 14 (19): 7751–65. doi:10.1093/nar/14.19.7751. PMC 311794. PMID 3022233.
  30. ^ McCarthy D, Minner C, Bernstein H, Bernstein C (October 1976). "DNA elongation rates and growing point distributions of wild-type phage T4 and a DNA-delay amber mutant". Journal of Molecular Biology. 106 (4): 963–81. doi:10.1016/0022-2836(76)90346-6. PMID 789903.
  31. ^ Mufti S, Bernstein H (October 1974). "The DNA-delay mutants of bacteriophage T4". Journal of Virology. 14 (4): 860–71. doi:10.1128/JVI.14.4.860-871.1974. PMC 355592. PMID 4609406.
  32. ^ Hyman P (August 1993). "The genetics of the Luria-Latarjet effect in bacteriophage T4: evidence for the involvement of multiple DNA repair pathways". Genetical Research. 62 (1): 1–9. doi:10.1017/s0016672300031499. PMID 8405988.
  33. ^ Miskimins R, Schneider S, Johns V, Bernstein H (June 1982). "Topoisomerase involvement in multiplicity reactivation of phage T4". Genetics. 101 (2): 157–77. doi:10.1093/genetics/101.2.157 (inactive 31 January 2024). PMC 1201854. PMID 6293912.{{cite journal}}: CS1 maint: DOI inactive as of January 2024 (link)
  34. ^ Cassuto E (September 1984). "Formation of covalently closed heteroduplex DNA by the combined action of gyrase and RecA protein". The EMBO Journal. 3 (9): 2159–64. doi:10.1002/j.1460-2075.1984.tb02106.x. PMC 557658. PMID 6092061.

Further reading edit

  • Wang JC (June 2002). "Cellular roles of DNA topoisomerases: a molecular perspective". Nature Reviews. Molecular Cell Biology. 3 (6): 430–40. doi:10.1038/nrm831. PMID 12042765. S2CID 205496065.

External links edit

type, topoisomerase, topoisomerases, that, both, strands, helix, simultaneously, order, manage, tangles, supercoils, they, hydrolysis, unlike, type, topoisomerase, this, process, these, enzymes, change, linking, number, circular, topoisomerases, ubiquitous, en. Type II topoisomerases are topoisomerases that cut both strands of the DNA helix simultaneously in order to manage DNA tangles and supercoils They use the hydrolysis of ATP unlike Type I topoisomerase In this process these enzymes change the linking number of circular DNA by 2 Topoisomerases are ubiquitous enzymes found in all living organisms 1 DNA Topoisomerase II ATP hydrolyzing Structure of the 42 KDa fragment of the N terminal ATPase and transducer domains of DNA gyrase homologous to all other type IIA topoisomerases IdentifiersEC no 5 6 2 2DatabasesIntEnzIntEnz viewBRENDABRENDA entryExPASyNiceZyme viewKEGGKEGG entryMetaCycmetabolic pathwayPRIAMprofilePDB structuresRCSB PDB PDBe PDBsumSearchPMCarticlesPubMedarticlesNCBIproteinsIn animals topoisomerase II is a chemotherapy target In prokaryotes gyrase is an antibacterial target 2 Indeed these enzymes are of interest for a wide range of effects Contents 1 Function 2 Topology simplification 3 Classification 4 Structure 4 1 Type IIA 4 2 Type IIB 5 Mechanism of action 5 1 Strand passage 5 2 DNA cleavage 6 Catenation 7 Inhibition 8 Bacteriophage T4 gyrase 9 References 10 Further reading 11 External linksFunction editType II topoisomerases increase or decrease the linking number of a DNA loop by 2 units and it promotes chromosome disentanglement For example DNA gyrase a type II topoisomerase observed in E coli and most other prokaryotes introduces negative supercoils and decreases the linking number by 2 Gyrase is also able to remove knots from the bacterial chromosome Along with gyrase most prokaryotes also contain a second type IIA topoisomerase termed topoisomerase IV Gyrase and topoisomerase IV differ by their C terminal domains which is believed to dictate substrate specificity and functionality for these two enzymes Footprinting indicates that gyrase which forms a 140 base pair footprint and wraps DNA introduces negative supercoils while topoisomerase IV which forms a 28 base pair footprint does not wrap DNA Eukaryotic type II topoisomerase cannot introduce supercoils it can only relax them The roles of type IIB topoisomerases are less understood Unlike type IIA topoisomerases type IIB topoisomerases cannot simplify DNA topology see below but they share several structural features with type IIA topoisomerases Topology simplification editType IIA topoisomerases are essential in the separation of entangled daughter strands during replication This function is believed to be performed by topoisomerase II in eukaryotes and by topoisomerase IV in prokaryotes Failure to separate these strands leads to cell death Type IIA topoisomerases have the special ability to relax DNA to a state below that of thermodynamic equilibrium a feature unlike type IA IB and IIB topoisomerases This ability known as topology simplification was first identified by Rybenkov et al 3 The hydrolysis of ATP drives this simplification but a clear molecular mechanism for this simplification is still lacking Several models to explain this phenomenon have been proposed including two models that rely on the ability of type IIA topoisomerases to recognize bent DNA duplexes 4 Biochemistry electron microscopy and recent structures of topoisomerase II bound to DNA reveal that type IIA topoisomerases bind at the apices of DNA supporting this model Classification editThis section is missing information about newly found IIB members doi 10 1093 nargab lqz021 Please expand the section to include this information Further details may exist on the talk page October 2021 There are two subclasses of type II topoisomerases type IIA and IIB Type IIA topoisomerases include the enzymes DNA gyrase eukaryotic topoisomerase II topo II and bacterial topoisomerase IV topo IV These enzymes span all domains of life and are essential for function 5 Type IIB topoisomerases are structurally and biochemically distinct and comprise a single family member topoisomerase VI topo VI Type IIB topoisomerases are found in archaea and some higher plants Some organisms including humans have two isoforms of topoisomerase II alpha and beta In cancers the topoisomerase IIa is highly expressed in proliferating cells In certain cancers such as peripheral nerve sheath tumors high expression of its encoded protein is also associated to poor patient survival The two classes of topoisomerases possess a similar strand passage mechanism and domain structure see below however they also have several important differences Type IIA topoisomerases form double stranded breaks with four base pair overhangs while type IIB topoisomerases form double stranded breaks with two base overhangs 6 In addition type IIA topoisomerases are able to simplify DNA topology 3 while type IIB topoisomerases do not 7 Structure editType IIA edit nbsp Schematic structure of gyrase oriented upside down compared to the other examples in this article nbsp Structure of yeast topoisomerase II bound to a doubly nicked 34 mer duplex DNA PDB 2RGR The Toprim fold is colored cyan the DNA is colored orange the HTH is colored magenta and the C gate is colored purple Notice that the DNA is bent by 160 degrees through an invariant isoleucine Ile833 in yeast Type IIA topoisomerases consist of several key motifs an N terminal GHKL ATPase domain for gyrase Hsp kinase and MutL a Toprim domain a Rossmann fold subclass which exists in both type II topoisomerases type IA topoisomerases and bacterial primase DnaG a central DNA binding core which structurally forms a heart shaped structure and a variable C terminal domain Eukaryotic type II topoisomerases are homodimers A2 while prokaryotic type II topoisomerases are heterotetramers A2B2 Prokaryotes have the ATPase domain and the Toprim fold on one polypeptide Pfam PF00204 while the DNA cleavage core and the CTD lies on a second polypeptide Pfam PF00521 For gyrase the first polypeptide is called GyrB and the second polypeptide is called GyrA For topo IV the first polypeptide is called ParE and the second polypeptide is called ParC Both Pfam signatures are found in the single chain eukayotic topoisomerase The structures of the N terminal ATPase domain of gyrase 8 and yeast topoisomerase II 9 have been solved in complex with AMPPNP an ATP analogue showing that two ATPase domains dimerize to form a closed conformation For gyrase the structure has a substantial hole in the middle which is presumed to accommodate the T segment Linking the ATPase domain to the Toprim fold is a helical element known as the transducer domain This domain is thought to communicate the nucleotide state of the ATPase domain to the rest of the protein Modifications to this domain affect topoisomerase activity and structural work done by the Verdine group shows that the ATP state affects the orientation of the transducer domain 10 The central core of the protein contains a Toprim fold and a DNA binding core that contains a winged helix domain WHD often referred to as a CAP domain since it was first identified to resemble the WHD of catabolite activator protein The catalytic tyrosine lies on this WHD The Toprim fold is a Rossmann fold that contains three invariant acidic residues that coordinate magnesium ions involved in DNA cleavage and DNA religation 11 The structure of the Toprim fold and DNA binding core of yeast topoisomerase II was first solved by Berger and Wang 12 and the first gyrase DNA binding core was solved by Morais Cabral et al 13 The structure solved by Berger revealed important insights into the function of the enzyme The DNA binding core consists of the WHD which leads to a tower domain A coiled coil region leads to a C terminal domain that forms the main dimer interface for this crystal state often termed the C gate While the original topoisomerase II structure shows a situation where the WHDs are separated by a large distance the structure of gyrase shows a closed conformation where the WHD close The topoisomerase II core was later solved in new conformations including one by Fass et al 14 and one by Dong et al 15 The Fass structure shows that the Toprim domain is flexible and that this flexibility can allow the Toprim domain to coordinate with the WHD to form a competent cleavage complex This was eventually substantiated by the Dong et al structure that was solved in the presence of DNA This last structure showed that the Toprim domain and the WHD formed a cleavage complex very similar to that of the type IA topoisomerases and indicated how DNA binding and cleavage could be uncoupled and the structure showed that DNA was bent by 150 degrees through an invariant isoleucine in topoisomerase II it is I833 and in gyrase it is I172 This mechanism of bending resembles closely that of integration host factor IHF and HU two architectural proteins in bacteria In addition while the previous structures of the DNA binding core had the C gate closed this structure captured the gate open a key step in the two gate mechanism see below More recently several structures of the DNA bound structure have been solved in an attempt to understand both the chemical mechanism for DNA cleavage and the structural basis for inhibition of topoisomerase by antibacterial poisons The first complete architecture of the E coli DNA gyrase has been solved by cryo electron microscopy at near atomic resolution 16 The nucleoprotein complex was captured with a long DNA duplex and gepotidacin a novel bacterial topoisomerase inhibitor The C terminal region of the prokaryotic topoisomerases has been solved for multiple species The first structure of a C terminal domain of gyrase was solved by Corbett et al 17 and the C terminal domain of topoisomerase IV was solved by Corbett et al 7 The structures formed a novel beta barrel which bends DNA by wrapping the nucleic acid around itself The bending of DNA by gyrase has been proposed as a key mechanism in the ability of gyrase to introduce negative supercoils into the DNA This is consistent with footprinting data that shows that gyrase has a 140 base pair footprint Both gyrase and topoisomerase IV CTDs bend DNA but only gyrase introduces negative supercoils Unlike the function of the C terminal domain of prokaryotic topoisomerases the function of the C terminal region of eukaryotic topoisomerase II is still not clear Studies have suggested that this region is regulated by phosphorylation and this modulates topoisomerase activity however more research needs to be done to investigate this Type IIB edit nbsp Structure of topo VI PDB 2Q2E in an orientation similar to the yeast example Chains are colored differently The Toprim domain lies on the top and the ATPase domain lies on the bottom each forms a DNA gate The organization of type IIB topoisomerases are similar to that of type IIAs except that all type IIBs have two genes and form heterotetramers One gene termed topo VI B since it resembles gyrB contains the ATPase domain a transducer domain Pfam PF09239 and a C terminal Ig fold like H2TH domain Pfam PF18000 The second gene termed topo VI A Pfam PF04406 contains the WHD and the Toprim domain The ATPase domain of topo VI B was solved in multiple nucleotide states 18 It closely resembles that of the GHKL domain of topo II and MutL and shows that the nucleotide state ADP versus ATP effects the orientation of the transducer domain and 1MX0 The structure of topo VI A was solved by Bergerat et al 19 showing that the HTH and Toprim fold had a novel conformation compared with that of topo IIA A recent structure of the topo VI A B complex was solved showing an open and closed conformation two states that are predicted in the two gate mechanism see below These structures of which one is an X ray crystal structure and the other is a Small Angle X ray Scattering SAXS reconstruction show that the ATPase domain can be either open or closed 20 Mechanism of action editStrand passage edit Type IIA topoisomerase operates through a two gate mechanism though this is a historical notation a mechanism supported by biochemistry 21 as well as by structural work 22 A strand of DNA called the gate or G segment is bound by a central DNA binding gate DNA gate A second strand of DNA called the transport or T segment is captured by the dimerization of the N terminal ATPase domain the ATPase gate when two molecules of ATP bind Hydrolysis of ATP and release of an inorganic phosphate leads to the cleavage of the G segment as the catalytic tyrosines form a covalent phosphotyrosine bond with the 5 end of the DNA This creates a four base overhang and a double stranded break in the G segment As the DNA binding gate separates the T segment is transferred through the G segment The G segment is sealed leading to the C terminal gate or C gate to open allowing for the release of the T segment Release of product ADP leads to a reset of the system and allows a second T segment to be captured Type IIB topoisomerases operate through a similar fashion except that the protein forms a two base overhang in the G segment and the C terminal gate is completely missing DNA cleavage edit In the strand passage mechanism the cleavage of DNA is key to allow the T segment to transfer through the G segment The mechanism of DNA cleavage by type IIA topoisomerases has recently been the focus of many biochemical and structural biology studies Catenation editCatenation is the process by which two circular DNA strands are linked together like chain links This occurs after DNA replication where two single strands are catenated and can still replicate but cannot separate into the two daughter cells As type II topoisomerses break a double strand they can fix this state type I topoisomerases could do this only if there were already a single strand nick and the correct chromosome number can remain in daughter cells Linear DNA in eukaryotes is so long they can be thought of as being without ends type II topoisomerases are needed for the same reason Inhibition editMain article Topoisomerase inhibitor Small molecules that target type II topoisomerase are divided into two classes inhibitors and poisons Due to their frequent presence in proliferating eukaryotic cells inhibitors of type II topoisomerases have been extensively studied and used as anti cancer medications 23 Inhibitors of type II topoisomerase include HU 331 ICRF 187 ICRF 193 and mitindomide These molecules work by inhibiting the ATPase activity by acting as noncompetitive inhibitors of ATP This has been shown through structural studies 9 and biochemical studies performed by the Lindsley group Poisons of type II topoisomerases include doxorubicin etoposide novobiocin quinolones including ciprofloxacin and teniposide These small molecules target the DNA protein complex Some of these molecules lead to increased cleavage whereas others such as etoposide inhibit religation The experimental antitumor drug m AMSA 4 9 acridinylamino methanesulfon m anisidide also inhibits type 2 topoisomerase 24 Topoisomerase poisons have been extensively used as both anticancer and antibacterial therapies While antibacterial compounds such as ciprofloxacin target bacterial gyrase they fail to inhibit eukaryotic type IIA topoisomerases In addition drug resistant bacteria often have a point mutation in gyrase Serine79Alanine in E coli that renders quinolones ineffective citation needed Recent structural studies have led to the discovery of a compound that no longer relies on this residue and therefore has efficacy against drug resistant bacteria citation needed Bacteriophage T4 gyrase editThe bacteriophage phage T4 gyrase type II topoismerase is a multisubunit protein consisting of the products of genes 39 52 and probably 60 25 26 It catalyses the relaxation of negatively or positively superhelical DNA and is employed in phage DNA replication during infection of the E coli bacterial host 27 The phage gene 52 protein shares homology with the E coli gyrase gyrA subunit 28 and the phage gene 39 protein shares homology with the gyr B subunit 29 Since the host E coli DNA gyrase can partially compensate for the loss of the phage T4 gene products mutants defective in either genes 39 52 or 60 do not completely abolish phage DNA replication but rather delay its initiation 27 The rate of DNA elongation is not slower than wild type in such mutant infections 30 Mutants defective in genes 39 52 or 60 show increased genetic recombination as well as increased base substitution and deletion mutation suggesting that the host compensated DNA synthesis is less accurate than that directed by wild type phage 31 A mutant defective in gene 39 shows increased sensitivity to inactivation by ultraviolet irradiation during the stage of phage infection after initiation of DNA replication when multiple copies of the phage chromosome are present 32 Mutants defective in genes 39 52 and 60 have reduced ability to carry out multiplicity reactivation a form of recombinational repair that can deal with different types of DNA damage 33 The gyrase specified by the genome of uninfected E coli also appears to participate in recombinational repair by providing an initiation point for the reciprocal strand exchange driven by the RecA protein 34 References edit Deweese JE Osheroff N February 2009 The DNA cleavage reaction of topoisomerase II wolf in sheep s clothing Nucleic Acids Research 37 3 738 748 doi 10 1093 nar gkn937 PMC 2647315 PMID 19042970 Reece RJ Maxwell A 1991 DNA gyrase structure and function Critical Reviews in Biochemistry and Molecular Biology 26 3 4 335 375 doi 10 3109 10409239109114072 PMID 1657531 a b Rybenkov VV Ullsperger C Vologodskii AV Cozzarelli NR August 1997 Simplification of DNA topology below equilibrium values by type II topoisomerases Science 277 5326 New York N Y 690 3 doi 10 1126 science 277 5326 690 PMID 9235892 Vologodskii AV Zhang W Rybenkov VV Podtelezhnikov AA Subramanian D Griffith JD Cozzarelli NR March 2001 Mechanism of topology simplification by type II DNA topoisomerases Proceedings of the National Academy of Sciences of the United States of America 98 6 3045 9 Bibcode 2001PNAS 98 3045V doi 10 1073 pnas 061029098 PMC 30604 PMID 11248029 Reece RJ Maxwell A January 1991 DNA gyrase structure and function Critical Reviews in Biochemistry and Molecular Biology 26 3 4 335 75 doi 10 3109 10409239109114072 PMID 1657531 Buhler C Lebbink JH Bocs C Ladenstein R Forterre P October 2001 DNA topoisomerase VI generates ATP dependent double strand breaks with two nucleotide overhangs The Journal of Biological Chemistry 276 40 37215 22 doi 10 1074 jbc M101823200 PMID 11485995 S2CID 24354635 a b PDB 1zvt Corbett KD Schoeffler AJ Thomsen ND Berger JM August 2005 The structural basis for substrate specificity in DNA topoisomerase IV Journal of Molecular Biology 351 3 545 61 doi 10 1016 j jmb 2005 06 029 PMID 16023670 Wigley DB Davies GJ Dodson EJ Maxwell A Dodson G June 1991 Crystal structure of an N terminal fragment of the DNA gyrase B protein Nature 351 6328 624 9 Bibcode 1991Natur 351 624W doi 10 1038 351624a0 PMID 1646964 S2CID 4373125 a b PDB 1PVG Classen S Olland S Berger JM September 2003 Structure of the topoisomerase II ATPase region and its mechanism of inhibition by the chemotherapeutic agent ICRF 187 Proceedings of the National Academy of Sciences of the United States of America 100 19 10629 34 Bibcode 2003PNAS 10010629C doi 10 1073 pnas 1832879100 PMC 196855 PMID 12963818 Wei H Ruthenburg AJ Bechis SK Verdine GL November 2005 Nucleotide dependent domain movement in the ATPase domain of a human type IIA DNA topoisomerase PDF The Journal of Biological Chemistry 280 44 37041 7 doi 10 1074 jbc M506520200 PMID 16100112 S2CID 35186716 Aravind L Leipe DD Koonin EV September 1998 Toprim a conserved catalytic domain in type IA and II topoisomerases DnaG type primases OLD family nucleases and RecR proteins Nucleic Acids Research 26 18 4205 13 doi 10 1093 nar 26 18 4205 PMC 147817 PMID 9722641 PDB 1BGW Berger JM Gamblin SJ Harrison SC Wang JC January 1996 Structure and mechanism of DNA topoisomerase II Nature 379 6562 225 32 Bibcode 1996Natur 379 225B doi 10 1038 379225a0 PMID 8538787 S2CID 4360011 PDB 1AB4 Morais Cabral JH Jackson AP Smith CV Shikotra N Maxwell A Liddington RC August 1997 Crystal structure of the breakage reunion domain of DNA gyrase Nature 388 6645 903 6 Bibcode 1997Natur 388 903M doi 10 1038 42294 PMID 9278055 S2CID 4320715 PDB 1BJT Fass D Bogden CE Berger JM April 1999 Quaternary changes in topoisomerase II may direct orthogonal movement of two DNA strands Nature Structural Biology 6 4 322 6 doi 10 1038 7556 PMID 10201398 S2CID 947461 PDB 2RGR Dong KC Berger JM December 2007 Structural basis for gate DNA recognition and bending by type IIA topoisomerases Nature 450 7173 1201 5 Bibcode 2007Natur 450 1201D doi 10 1038 nature06396 PMID 18097402 S2CID 1756317 Vanden Broeck A Lotz C Ortiz J Lamour V October 2019 Cryo EM structure of the complete E coli DNA gyrase nucleoprotein complex Nature Communications 10 1 4935 Bibcode 2019NatCo 10 4935V doi 10 1038 s41467 019 12914 y PMC 6821735 PMID 31666516 PDB 1SUU Corbett KD Shultzaberger RK Berger JM May 2004 The C terminal domain of DNA gyrase A adopts a DNA bending beta pinwheel fold Proceedings of the National Academy of Sciences of the United States of America 101 19 7293 8 Bibcode 2004PNAS 101 7293C doi 10 1073 pnas 0401595101 PMC 409912 PMID 15123801 PDB 1MU5 Corbett KD Berger JM January 2003 Structure of the topoisomerase VI B subunit implications for type II topoisomerase mechanism and evolution The EMBO Journal 22 1 151 63 doi 10 1093 emboj cdg008 PMC 140052 PMID 12505993 Bergerat A de Massy B Gadelle D Varoutas PC Nicolas A Forterre P March 1997 An atypical topoisomerase II from Archaea with implications for meiotic recombination Nature 386 6623 414 7 Bibcode 1997Natur 386 414B doi 10 1038 386414a0 PMID 9121560 S2CID 4327493 PDB 2Q2E Corbett KD Benedetti P Berger JM July 2007 Holoenzyme assembly and ATP mediated conformational dynamics of topoisomerase VI Nature Structural amp Molecular Biology 14 7 611 9 doi 10 1038 nsmb1264 PMID 17603498 S2CID 2159631 Roca J Wang JC May 1994 DNA transport by a type II DNA topoisomerase evidence in favor of a two gate mechanism Cell 77 4 609 16 doi 10 1016 0092 8674 94 90222 4 PMID 8187179 S2CID 19776252 Berger JM Wang JC February 1996 Recent developments in DNA topoisomerase II structure and mechanism Current Opinion in Structural Biology 6 1 84 90 doi 10 1016 s0959 440x 96 80099 6 PMID 8696977 Alberts B 2014 11 18 Molecular biology of the cell Sixth ed New York NY ISBN 978 0 8153 4432 2 OCLC 887605755 a href Template Cite book html title Template Cite book cite book a CS1 maint location missing publisher link Willmore E de Caux S Sunter NJ Tilby MJ Jackson GH Austin CA Durkacz BW June 2004 A novel DNA dependent protein kinase inhibitor NU7026 potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia Blood 103 12 4659 65 doi 10 1182 blood 2003 07 2527 PMID 15010369 Liu LF Liu CC Alberts BM October 1979 T4 DNA topoisomerase a new ATP dependent enzyme essential for initiation of T4 bacteriophage DNA replication Nature 281 5731 456 61 Bibcode 1979Natur 281 456L doi 10 1038 281456a0 PMID 226889 S2CID 4343962 Stetler GL King GJ Huang WM August 1979 T4 DNA delay proteins required for specific DNA replication form a complex that has ATP dependent DNA topoisomerase activity Proceedings of the National Academy of Sciences of the United States of America 76 8 3737 41 Bibcode 1979PNAS 76 3737S doi 10 1073 pnas 76 8 3737 PMC 383908 PMID 226976 a b McCarthy D January 1979 Gyrase dependent initiation of bacteriophage T4 DNA replication interactions of Escherichia coli gyrase with novobiocin coumermycin and phage DNA delay gene products Journal of Molecular Biology 127 3 265 83 doi 10 1016 0022 2836 79 90329 2 PMID 372540 Huang WM September 1986 The 52 protein subunit of T4 DNA topoisomerase is homologous to the gyrA protein of gyrase Nucleic Acids Research 14 18 7379 90 PMC 311757 PMID 3020513 Huang WM October 1986 Nucleotide sequence of a type II DNA topoisomerase gene Bacteriophage T4 gene 39 Nucleic Acids Research 14 19 7751 65 doi 10 1093 nar 14 19 7751 PMC 311794 PMID 3022233 McCarthy D Minner C Bernstein H Bernstein C October 1976 DNA elongation rates and growing point distributions of wild type phage T4 and a DNA delay amber mutant Journal of Molecular Biology 106 4 963 81 doi 10 1016 0022 2836 76 90346 6 PMID 789903 Mufti S Bernstein H October 1974 The DNA delay mutants of bacteriophage T4 Journal of Virology 14 4 860 71 doi 10 1128 JVI 14 4 860 871 1974 PMC 355592 PMID 4609406 Hyman P August 1993 The genetics of the Luria Latarjet effect in bacteriophage T4 evidence for the involvement of multiple DNA repair pathways Genetical Research 62 1 1 9 doi 10 1017 s0016672300031499 PMID 8405988 Miskimins R Schneider S Johns V Bernstein H June 1982 Topoisomerase involvement in multiplicity reactivation of phage T4 Genetics 101 2 157 77 doi 10 1093 genetics 101 2 157 inactive 31 January 2024 PMC 1201854 PMID 6293912 a href Template Cite journal html title Template Cite journal cite journal a CS1 maint DOI inactive as of January 2024 link Cassuto E September 1984 Formation of covalently closed heteroduplex DNA by the combined action of gyrase and RecA protein The EMBO Journal 3 9 2159 64 doi 10 1002 j 1460 2075 1984 tb02106 x PMC 557658 PMID 6092061 Further reading editWang JC June 2002 Cellular roles of DNA topoisomerases a molecular perspective Nature Reviews Molecular Cell Biology 3 6 430 40 doi 10 1038 nrm831 PMID 12042765 S2CID 205496065 External links edit nbsp Wikimedia Commons has media related to Type II DNA topoisomerase DNA Topoisomerases Type II at the U S National Library of Medicine Medical Subject Headings MeSH Portal nbsp Biology Retrieved from https en wikipedia org w index php title Type II topoisomerase amp oldid 1203793970 Inhibition, 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.