ERCC6

Last updated
ERCC6
ERCC6 protein.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases ERCC6 , ARMD5, CKN2, COFS, COFS1, CSB, RAD26, UVSS1, POF11, ERCC excision repair 6, chromatin remodeling factor, excision repair cross-complementing group 6
External IDs OMIM: 609413 MGI: 1100494 HomoloGene: 133552 GeneCards: ERCC6
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

NM_001081221

RefSeq (protein)

NP_000115
NP_001263987
NP_001263988
NP_001333369
NP_736609

Contents

NP_001074690

Location (UCSC)n/a Chr 14: 32.24 – 32.3 Mb
PubMed search [2] [3]
Wikidata
View/Edit Human View/Edit Mouse

DNA excision repair protein ERCC-6 (also CS-B protein) is a protein that in humans is encoded by the ERCC6 gene. [4] [5] [6] The ERCC6 gene is located on the long arm of chromosome 10 at position 11.23. [7]

Having 1 or more copies of a mutated ERCC6 causes Cockayne syndrome, type II.

Function

DNA can be damaged by ultraviolet radiation, toxins, radioactive substances, and reactive biochemical intermediates like free radicals. The ERCC6 protein is involved in repairing the genome when specific genes undergoing transcription (dubbed active genes) are inoperative; as such, ERCC6 serves as a transcription-coupled excision repair protein, being one of the fundamental enzymes in active gene repair. [7]

Structure and Mechanism

CSB has been found to exhibit ATPase properties; there are contradictory publications regarding the effect of ATP concentration on CSB's activity. [8] The most recent evidence suggests that ADP/AMP allosterically regulate CSB. [6] As such, it has been speculated that CSB may promote protein complex formation at repair sites subject to the ATP to ADP charge ratio.

Conservation of helicase motifs in eukaryote CSB is evident; all seven major domains of the protein are conserved among numerous RNA and DNA helicases. Detailed structural analysis of CSB has been performed; motifs I, Ia, II, and III are collectively called domain 1, while motifs IV, V, and VI comprise domain 2. These domains wrap around an interdomain cleft involved in ATP binding and hydrolysis. Motifs III and IV are in close proximity to the active site; hence, residues in these regions stabilize ATP/ADP binding via hydrogen bonding. [9] Domain 2 has been proposed to affect DNA binding after induced conformational changes stemming from ATP hydrolysis. Specific residues involved in gene binding have yet to be identified. [10]

The evolutionary roots of CSB has led some to contend that it exhibits helicase activity. [11] Evidence for the helicase properties of CSB is highly disputed; yet, it has been found the protein participates in intracellular trafficking, a traditional role of helicases. The complex interactions between DNA repair proteins suggest that eukaryote CSB upholds some but not all of the functions of its prokaryotic precursors. [12]

Interactions

CSB has been shown to interact with P53. [13] [14]

CSB has been shown to act as chromatin remodeling factor for RNA Polymerase II. When RNA Polymerase II is stalled by a mistake in the genome, CSB remodels the DNA double helix so as to allow access by repair enzymes to the lesion. [15]

CSB is involved in the base excision repair (BER) pathway. This is due to demonstrated interactions with human AP endonuclease, though interactions between recombinant CSB and E. coli endonuclease IV as well as human N-terminus AP endonuclease fragments have not been observed in vitro. Specifically, CSB stimulates the AP site incision activity of AP endonuclease independent of ATP. [16]

In addition to the BER pathway, CSB is heavily integrated in the nucleotide excision repair (NER) pathway. While BER utilizes glycosylases to recognize and correct non-bulky lesions, NER is particularly versatile in repairing DNA damaged by UV radiation via the removal of oxidized bases. CSB's role in NER is best manifested by interactions with T cell receptors, in which protein collaboration is key in effective antigen binding. [17]

Neurogenesis and Neural Differentiation

ERCC6 knockout within human neural progenitor cells has been shown to decrease both neurogenesis and neural differentiation. Both mechanisms are key in brain development, explaining characteristic cognitive deficiencies of Cockayne syndrome - such as stunted development of the nervous system - that otherwise do not seem related to symptoms like photosensitivity and hearing loss. [18]

Cockayne syndrome

In humans, Cockayne syndrome (CS) is a rare autosomal recessive leukodystrophy (associated with the degradation of white matter). CS arises from germ line mutations in either of two genes, CSA(ERCC8) or CSB(ERCC6). About two thirds of CS patients have a mutation in the CSB(ERCC6) gene. [19] Mutations in ERCC6 that lead to CS deal with both the size of the protein as well as the specific amino acid residues utilized in biosynthesis. Patients exhibiting type II CS often have shortened and/or misfolded CSB that disrupt gene expression and transcription. The characteristic biological effect of malfunctioning ERCC6 is nerve cell death, resulting in premature aging and growth defects. [7]

The extent to which malfunctioning CSB hinders oxidative repair heavily influences patients' neurological functioning. The two subforms of the disorder (the latter of which corresponds to ERCC6 defects) - CS-A and CS-B - both cause problems in the oxidative repair, though CS-B patients more often exhibit nerve system problems stemming from damage to this pathway. Most type II CS patients exhibit photosensitivity as per the heavily oxidative properties of UV light. [20] [21]

While two copies of mutated ERCC6 result in CS, possession of a single copy of mutated ERCC6 gene is associated with similar but milder defects as CS, including retinal dystrophy, cardiac arrhythmias, and immunodeficiency. [22] Individuals who are heterozygote carriers are therefore at increased risk of similar pleiotropic disorders as homozygote carriers afflicted with CS.

DNA repair

CSB and CSA proteins are considered to function in transcription coupled nucleotide excision repair (TC-NER). CSB and CSA deficient cells are unable to preferentially repair UV-induced cyclobutane pyrimidine dimers in actively transcribed genes, consistent with a failed TC-NER response. [23] CSB also accumulates at sites of DNA double-strand breaks in a transcription dependent manner and influences double-strand break repair. [24] CSB protein facilitates homologous recombinational repair of double-strand breaks and represses non-homologous end joining. [24]

In a damaged cell, the CSB protein localizes to sites of DNA damage. CSB recruitment to damaged sites is influenced by the type of DNA damage and is, most rapid and robust as follows: interstrand crosslinks > double-strand breaks > monoadducts > oxidative damages. [19] The CSB protein interacts with SNM1A(DCLRE1A) protein, a 5’- 3’ exonuclease, to promote the removal of DNA interstrand crosslinks. [25]

Implications in cancer

Single-nucleotide polymorphisms in the ERCC6 gene have been correlated with significantly increased risk of certain forms of cancer. A specific mutation at the 1097 position (M1097V) as well as polymorphisms at amino acid residue 1413 have been associated with heightened risk of bladder cancer for experimental subjects in Taiwan; moreover, M1097V has been argued to play a key role in pathogenesis. [26] Rs1917799 polymorphism has been associated with increased risk of gastric cancer for Chinese experimental subjects, [27] and mutations at codon 399 have been correlated to the onset of oral cancers among Taiwanese patients. [28] Another study found a diverse set of mutations in the ERCC6 gene among Chinese lung cancer patients versus the general population (in terms of statistical significance), but failed to identify specific polymorphisms correlated with the patients' illness. [29]

Faulty DNA repair is implicated causally in tumor development due to malfunctioning proteins' inability to correct genes responsible for apoptosis and cell growth. Yet, the vast majority of studies regarding the effects of ERCC6 knockout or mutations on cancer are based upon statistical correlations of available patient data as opposed to mechanistic analysis of in vivo cancer onset. Hence, confounding based on protein-protein, protein-substrate, and/or substrate-substrate interactions disallows conclusions positing mutations in ERCC6 cause cancer on an individual basis.

Related Research Articles

<span class="mw-page-title-main">Werner syndrome</span> Medical condition

Werner syndrome (WS) or Werner's syndrome, also known as "adult progeria", is a rare, autosomal recessive disorder which is characterized by the appearance of premature aging.

<span class="mw-page-title-main">Helicase</span> Class of enzymes to unpack an organisms genes

Helicases are a class of enzymes thought to be vital to all organisms. Their main function is to unpack an organism's genetic material. Helicases are motor proteins that move directionally along a nucleic acid phosphodiester backbone, separating two hybridized nucleic acid strands, using energy from ATP hydrolysis. There are many helicases, representing the great variety of processes in which strand separation must be catalyzed. Approximately 1% of eukaryotic genes code for helicases.

<span class="mw-page-title-main">Xeroderma pigmentosum</span> Medical condition

Xeroderma pigmentosum (XP) is a genetic disorder in which there is a decreased ability to repair DNA damage such as that caused by ultraviolet (UV) light. Symptoms may include a severe sunburn after only a few minutes in the sun, freckling in sun-exposed areas, dry skin and changes in skin pigmentation. Nervous system problems, such as hearing loss, poor coordination, loss of intellectual function and seizures, may also occur. Complications include a high risk of skin cancer, with about half having skin cancer by age 10 without preventative efforts, and cataracts. There may be a higher risk of other cancers such as brain cancers.

<span class="mw-page-title-main">Cockayne syndrome</span> Medical condition

Cockayne syndrome (CS), also called Neill-Dingwall syndrome, is a rare and fatal autosomal recessive neurodegenerative disorder characterized by growth failure, impaired development of the nervous system, abnormal sensitivity to sunlight (photosensitivity), eye disorders and premature aging. Failure to thrive and neurological disorders are criteria for diagnosis, while photosensitivity, hearing loss, eye abnormalities, and cavities are other very common features. Problems with any or all of the internal organs are possible. It is associated with a group of disorders called leukodystrophies, which are conditions characterized by degradation of neurological white matter. The underlying disorder is a defect in a DNA repair mechanism. Unlike other defects of DNA repair, patients with CS are not predisposed to cancer or infection. Cockayne syndrome is a rare but destructive disease usually resulting in death within the first or second decade of life. The mutation of specific genes in Cockayne syndrome is known, but the widespread effects and its relationship with DNA repair is yet to be well understood.

<span class="mw-page-title-main">Nucleotide excision repair</span> DNA repair mechanism

Nucleotide excision repair is a DNA repair mechanism. DNA damage occurs constantly because of chemicals, radiation and other mutagens. Three excision repair pathways exist to repair single stranded DNA damage: Nucleotide excision repair (NER), base excision repair (BER), and DNA mismatch repair (MMR). While the BER pathway can recognize specific non-bulky lesions in DNA, it can correct only damaged bases that are removed by specific glycosylases. Similarly, the MMR pathway only targets mismatched Watson-Crick base pairs.

<span class="mw-page-title-main">XPB</span> Mammalian protein found in Homo sapiens

XPB is an ATP-dependent DNA helicase in humans that is a part of the TFIIH transcription factor complex.

A DNA repair-deficiency disorder is a medical condition due to reduced functionality of DNA repair.

<span class="mw-page-title-main">ERCC2</span> Mammalian protein found in humans

ERCC2, or XPD is a protein involved in transcription-coupled nucleotide excision repair.

Transcription factor II Human is an important protein complex, having roles in transcription of various protein-coding genes and DNA nucleotide excision repair (NER) pathways. TFIIH first came to light in 1989 when general transcription factor-δ or basic transcription factor 2 was characterized as an indispensable transcription factor in vitro. This factor was also isolated from yeast and finally named as TFIIH in 1992.

<span class="mw-page-title-main">ERCC1</span> Protein-coding gene in the species Homo sapiens

DNA excision repair protein ERCC-1 is a protein that in humans is encoded by the ERCC1 gene. Together with ERCC4, ERCC1 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination.

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

DNA repair protein complementing XP-A cells is a protein that in humans is encoded by the XPA gene.

<span class="mw-page-title-main">ERCC5</span>

DNA repair protein complementing XP-G cells is a protein that in humans is encoded by the ERCC5 gene.

<span class="mw-page-title-main">ERCC4</span> Protein-coding gene in the species Homo sapiens

ERCC4 is a protein designated as DNA repair endonuclease XPF that in humans is encoded by the ERCC4 gene. Together with ERCC1, ERCC4 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination.

<span class="mw-page-title-main">ERCC8 (gene)</span> Protein-coding gene in the species Homo sapiens

DNA excision repair protein ERCC-8 is a protein that in humans is encoded by the ERCC8 gene.

<span class="mw-page-title-main">GTF2H2</span> Protein-coding gene in the species Homo sapiens

General transcription factor IIH subunit 2 is a protein that in humans is encoded by the GTF2H2 gene.

<span class="mw-page-title-main">GTF2H1</span> Protein-coding gene in the species Homo sapiens

General transcription factor IIH subunit 1 is a protein that in humans is encoded by the GTF2H1 gene.

RNA polymerase II holoenzyme is a form of eukaryotic RNA polymerase II that is recruited to the promoters of protein-coding genes in living cells. It consists of RNA polymerase II, a subset of general transcription factors, and regulatory proteins known as SRB proteins.

The DNA damage theory of aging proposes that aging is a consequence of unrepaired accumulation of naturally occurring DNA damage. Damage in this context is a DNA alteration that has an abnormal structure. Although both mitochondrial and nuclear DNA damage can contribute to aging, nuclear DNA is the main subject of this analysis. Nuclear DNA damage can contribute to aging either indirectly or directly.

Genome instability refers to a high frequency of mutations within the genome of a cellular lineage. These mutations can include changes in nucleic acid sequences, chromosomal rearrangements or aneuploidy. Genome instability does occur in bacteria. In multicellular organisms genome instability is central to carcinogenesis, and in humans it is also a factor in some neurodegenerative diseases such as amyotrophic lateral sclerosis or the neuromuscular disease myotonic dystrophy.

Progeroid syndromes (PS) are a group of rare genetic disorders that mimic physiological aging, making affected individuals appear to be older than they are. The term progeroid syndrome does not necessarily imply progeria, which is a specific type of progeroid syndrome.

References

  1. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000054051 - Ensembl, May 2017
  2. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  3. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. Troelstra C, van Gool A, de Wit J, Vermeulen W, Bootsma D, Hoeijmakers JH (Dec 1992). "ERCC6, a member of a subfamily of putative helicases, is involved in Cockayne's syndrome and preferential repair of active genes". Cell. 71 (6): 939–53. doi:10.1016/0092-8674(92)90390-X. hdl: 1765/3041 . PMID   1339317. S2CID   30671008.
  5. Muftuoglu M, de Souza-Pinto NC, Dogan A, Aamann M, Stevnsner T, Rybanska I, Kirkali G, Dizdaroglu M, Bohr VA (Apr 2009). "Cockayne syndrome group B protein stimulates repair of formamidopyrimidines by NEIL1 DNA glycosylase". The Journal of Biological Chemistry. 284 (14): 9270–9. doi: 10.1074/jbc.M807006200 . PMC   2666579 . PMID   19179336.
  6. 1 2 "Entrez Gene: ERCC6 excision repair cross-complementing rodent repair deficiency, complementation group 6".
  7. 1 2 3 NIH. "ERCC6 Gene." Genetics Home Reference. National Institutes of Health, 16 Feb. 2015. Web. 22 Feb. 2015. <http://ghr.nlm.nih.gov/gene/ERCC6>.
  8. Selby CP, Sancar A (Jan 17, 1997). "Human transcription-repair coupling factor CSB/CSB is a DNA-stimulated ATPase but is not a helicase and does not disrupt the ternary transcription complex of stalled RNA polymerase II". J Biol Chem. 272 (3): 1885–90. doi: 10.1074/jbc.272.3.1885 . PMID   8999876.
  9. Durr H, Korner C, Muller M, Hickmann V, Hopfner KP (2005). "X-ray structures of the Sulfolobus solfataricus SWI2/SNF2 ATPase core and its complex with DNA". Cell. 121 (3): 363–373. doi: 10.1016/j.cell.2005.03.026 . PMID   15882619.
  10. Lewis R, Durr H, Hopfner KP, Michaelis J (2008). "Conformational changes of a Swi2/ Snf2 ATPase during its mechano-chemical cycle". Nucleic Acids Res. 36 (6): 1881–1890. doi:10.1093/nar/gkn040. PMC   2346605 . PMID   18267970.
  11. Troelstra C, van Gool A, de Wit J, Vermeulen W, Bootsma D, Hoeijmakers JH (1993). "CSB, a member of a subfamily of putative helicases, is involved in Cockayne's syndrome and preferential repair of active genes". Cell. 71 (6): 939–53. doi:10.1016/0092-8674(92)90390-x. hdl: 1765/3041 . PMID   1339317. S2CID   30671008.
  12. Boulikas, T (March–April 1997). "Nuclear import of DNA repair proteins". Anticancer Research. 17 (2A): 843–63. PMID   9137418.
  13. Wang XW, Yeh H, Schaeffer L, Roy R, Moncollin V, Egly JM, Wang Z, Freidberg EC, Evans MK, Taffe BG (Jun 1995). "p53 modulation of TFIIH-associated nucleotide excision repair activity". Nature Genetics. 10 (2): 188–95. doi:10.1038/ng0695-188. hdl: 1765/54884 . PMID   7663514. S2CID   38325851.
  14. Yu A, Fan HY, Liao D, Bailey AD, Weiner AM (May 2000). "Activation of p53 or loss of the Cockayne syndrome group B repair protein causes metaphase fragility of human U1, U2, and 5S genes". Molecular Cell. 5 (5): 801–10. doi: 10.1016/S1097-2765(00)80320-2 . PMID   10882116.
  15. Newman JC, Bailey AD, Weiner AM (Jun 2006). "Cockayne syndrome group B protein (CSB) plays a general role in chromatin maintenance and remodeling". Proceedings of the National Academy of Sciences of the United States of America. 103 (25): 9313–8. Bibcode:2006PNAS..103.9613N. doi: 10.1073/pnas.0510909103 . PMC   1480455 . PMID   16772382.
  16. Wong HK, Muftuoglu M, Beck G, Imam SZ, Bohr VA, Wilson DM (June 2007). "Cockayne syndrome B protein stimulates apurinic endonuclease 1 activity and protects against agents that introduce base excision repair intermediates". Nucleic Acids Research. 35 (12): 4103–13. doi:10.1093/nar/gkm404. PMC   1919475 . PMID   17567611.
  17. Frosina G (Jul 2007). "The current evidence for defective repair of oxidatively damaged DNA in Cockayne syndrome". Free Radical Biology & Medicine. 43 (2): 165–77. doi:10.1016/j.freeradbiomed.2007.04.001. PMID   17603927.
  18. Ciaffardini F, Nicolai S, Caputo M, Canu G, Paccosi E, Costantino M, Frontini M, Balajee AS, Proietti-De-Santis L (2014). "The cockayne syndrome B protein is essential for neuronal differentiation and neuritogenesis". Cell Death Dis. 5 (5): e1268. doi:10.1038/cddis.2014.228. PMC   4047889 . PMID   24874740.
  19. 1 2 Iyama T, Wilson DM (2016). "Elements That Regulate the DNA Damage Response of Proteins Defective in Cockayne Syndrome". J. Mol. Biol. 428 (1): 62–78. doi:10.1016/j.jmb.2015.11.020. PMC   4738086 . PMID   26616585.
  20. Laugel, V., C. Dalloz, M. Durrand, and H. Dollfus. "Mutation Update for the CSB/ERCC6 and CSA/ERCC8 Genes Involved in Cockayne Syndrome." Human Mutation. Human Genome Variation Society, 5 Nov. 2009. Web. 22 Feb. 2015. <http://onlinelibrary.wiley.com/doi/10.1002/humu.21154/epdf>.
  21. Nardo T, Oneda R, Spivak G, Mortier L, Thomas P, Orioli D, Laugel V, Stary A, Hanawalt PC, Sarasin A, Stefanini M (2009). "A UV-sensitive syndrome patient with a specific CSA mutation reveals separable roles for CSA in response to UV and oxidative DNA damage". Proc Natl Acad Sci USA. 106 (15): 6209–6214. Bibcode:2009PNAS..106.6209N. doi: 10.1073/pnas.0902113106 . PMC   2667150 . PMID   19329487.
  22. Forrest, Iain S.; Chaudhary, Kumardeep; Vy, Ha My T.; Bafna, Shantanu; Kim, Soyeon; Won, Hong-Hee; Loos, Ruth J. F.; Cho, Judy; Pasquale, Louis R.; Nadkarni, Girish N.; Rocheleau, Ghislain (2021). "Genetic pleiotropy of ERCC6 loss-of-function and deleterious missense variants links retinal dystrophy, arrhythmia, and immunodeficiency in diverse ancestries". Human Mutation. 42 (8): 969–977. doi: 10.1002/humu.24220 . ISSN   1098-1004. PMC   8295228 . PMID   34005834.
  23. van Hoffen A, Natarajan AT, Mayne LV, van Zeeland AA, Mullenders LH, Venema J (1993). "Deficient repair of the transcribed strand of active genes in Cockayne's syndrome cells". Nucleic Acids Res. 21 (25): 5890–5. doi:10.1093/nar/21.25.5890. PMC   310470 . PMID   8290349.
  24. 1 2 Batenburg NL, Thompson EL, Hendrickson EA, Zhu XD (2015). "Cockayne syndrome group B protein regulates DNA double-strand break repair and checkpoint activation". EMBO J. 34 (10): 1399–416. doi:10.15252/embj.201490041. PMC   4491999 . PMID   25820262.
  25. Iyama T, Lee SY, Berquist BR, Gileadi O, Bohr VA, Seidman MM, McHugh PJ, Wilson DM (2015). "CSB interacts with SNM1A and promotes DNA interstrand crosslink processing". Nucleic Acids Res. 43 (1): 247–58. doi:10.1093/nar/gku1279. PMC   4288174 . PMID   25505141.
  26. Chang CH, Chiu CF, Wang HC, Wu HC, Tsai RY, Tsai CW, Wang RF, Wang CH, Tsou YA, Bau DT (2009). "Significant association of ERCC6 single nucleotide polymorphisms with bladder cancer susceptibility in Taiwan". Anticancer Res. 29 (12): 5121–4. PMID   20044625.
  27. Liu JW, He CY, Sun LP, Xu Q, Xing CZ, Yuan Y (2013). "The DNA repair gene ERCC6 rs1917799 polymorphism is associated with gastric cancer risk in Chinese". Asian Pac. J. Cancer Prev. 14 (10): 6103–8. doi: 10.7314/apjcp.2013.14.10.6103 . PMID   24289633.
  28. Chiu CF, Tsai MH, Tseng HC, Wang CL, Tsai FJ, Lin CC, Bau DT (2008). "A novel single nucleotide polymorphism in ERCC6 gene is associated with oral cancer susceptibility in Taiwanese patients". Oral Oncol. 44 (6): 582–6. doi:10.1016/j.oraloncology.2007.07.006. PMID   17933579.
  29. Ma H, Hu Z, Wang H, Jin G, Wang Y, Sun W, Chen D, Tian T, Jin L, Wei Q, Lu D, Huang W, Shen H (2009). "ERCC6/CSB gene polymorphisms and lung cancer risk". Cancer Lett. 273 (1): 172–6. doi:10.1016/j.canlet.2008.08.002. PMID   18789574.

Further reading