ERCC1

Last updated
ERCC1
Protein ERCC1 PDB 1z00.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases ERCC1 , COFS4, RAD10, UV20, excision repair cross-complementation group 1, ERCC excision repair 1, endonuclease non-catalytic subunit
External IDs OMIM: 126380 MGI: 95412 HomoloGene: 1501 GeneCards: ERCC1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001166049
NM_001983
NM_202001

NM_001127324
NM_007948

RefSeq (protein)

NP_001120796
NP_031974

Location (UCSC) Chr 19: 45.41 – 45.48 Mb Chr 7: 19.08 – 19.09 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

DNA excision repair protein ERCC-1 is a protein that in humans is encoded by the ERCC1 gene. [5] Together with ERCC4, ERCC1 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination. [6] [7]

Many aspects of these two gene products are described together here because they are partners during DNA repair. The ERCC1-XPF nuclease is an essential activity in the pathway of DNA nucleotide excision repair (NER). The ERCC1-XPF nuclease also functions in pathways to repair double-strand breaks in DNA, and in the repair of “crosslink” damage that harmfully links the two DNA strands.

Cells with disabling mutations in ERCC1 are more sensitive than normal to particular DNA damaging agents, including ultraviolet (UV) radiation and to chemicals that cause crosslinking between DNA strands. Genetically engineered mice with disabling mutations in ERCC1 have defects in DNA repair, accompanied by metabolic stress-induced changes in physiology that result in premature aging. [8] Complete deletion of ERCC1 is incompatible with viability of mice, and no human individuals have been found with complete (homozygous) deletion of ERCC1. Rare individuals in the human population harbor inherited mutations that impair the function of ERCC1. When the normal genes are absent, these mutations can lead to human syndromes, including Cockayne syndrome (CS) and COFS.

ERCC1 and ERCC4 are the gene names assigned in mammalian genomes, including the human genome (Homo sapiens). Similar genes with similar functions are found in all eukaryotic organisms.

Gene

The genomic DNA for ERCC1 was the first human DNA repair gene to be isolated by molecular cloning. The original method was by transfer of fragments of the human genome to ultraviolet light (UV)-sensitive mutant cell lines derived from Chinese hamster ovary cells. [9] Reflecting this cross-species genetic complementation method, the gene was called “Excision repair cross-complementing 1”. Multiple independent complementation groups of Chinese hamster ovary (CHO) cells were isolated, [10] and this gene restored UV resistance to cells of complementation group 1.

The human ERCC1 gene encodes the ERCC1 protein of 297 amino acids with a molecular mass of about 32,500 daltons.

Genes similar to ERCC1 with equivalent functions (orthologs) are found in other eukaryotic genomes. Some of the most studied gene orthologs include RAD10 in the budding yeast Saccharomyces cerevisiae , and swi10+ in the fission yeast Schizosaccharomyces pombe .

Protein

Diagram of ERCC1 showing a central domain and a helix-hairpin-helix domain Fig 1 ERCC1 Wikigene.png
Diagram of ERCC1 showing a central domain and a helix-hairpin-helix domain

One ERCC1 molecule and one XPF molecule bind together, forming an ERCC1-XPF heterodimer which is the active nuclease form of the enzyme. In the ERCC1–XPF heterodimer, ERCC1 mediates DNA– and protein–protein interactions. XPF provides the endonuclease active site and is involved in DNA binding and additional protein–protein interactions. [9]

The ERCC4/XPF protein consists of two conserved domains separated by a less conserved region in the middle. The N-terminal region has homology to several conserved domains of DNA helicases belonging to superfamily II, although XPF is not a DNA helicase. [11] The C-terminal region of XPF includes the active site residues for nuclease activity. [12] Most of the ERCC1 protein is related at the sequence level to the C-terminus of the XPF protein, [13] but residues in the nuclease domain are not present. A DNA binding “helix-hairpin-helix” domain at the C-terminus of each protein.

By primary sequence and protein structural similarity, the ERCC1-XPF nuclease is a member of a broader family of structure specific DNA nucleases comprising two subunits. Such nucleases include, for example, the MUS81-EME1 nuclease.

Structure-specific nuclease

DNA substrates of ERCC1-XPF nuclease Fig 2 XPF Substrate Wikigene.png
DNA substrates of ERCC1-XPF nuclease

The ERCC1–XPF complex is a structure-specific endonuclease. ERCC1-XPF does not cut DNA that is exclusively single-stranded or double-stranded, but it cleaves the DNA phosphodiester backbone specifically at junctions between double-stranded and single-stranded DNA. It introduces a cut in double-stranded DNA on the 5′ side of such a junction, about two nucleotides away. [14] This structure-specificity was initially demonstrated for RAD10-RAD1, the yeast orthologs of ERCC1 and XPF. [15]

The hydrophobic helix–hairpin–helix motifs in the C-terminal regions of ERCC1 and XPF interact to promote dimerization of the two proteins. [16] There is no catalytic activity in the absence of dimerization. Indeed, although the catalytic domain is within XPF and ERCC1 is catalytically inactive, ERCC1 is indispensable for activity of the complex.

Several models have been proposed for binding of ERCC1–XPF to DNA, based on partial structures of relevant protein fragments at atomic resolution. [16] DNA binding mediated by the helix-hairpin-helix domains of ERCC1 and XPF domains positions the heterodimer at the junction between double-stranded and single-stranded DNA.

Nucleotide excision repair

During nucleotide excision repair, several protein complexes cooperate to recognize damaged DNA and locally separate the DNA helix for a short distance on either side of the site of a DNA damage. The ERCC1–XPF nuclease incises the damaged DNA strand on the 5′ side of the lesion. [14] During NER, the ERCC1 protein interacts with the XPA protein to coordinate DNA and protein binding.

DNA double-strand break repair

Mammalian cells with mutant ERCC1–XPF are moderately more sensitive than normal cells to agents (such as ionizing radiation) that cause double-stranded breaks in DNA. [17] [18] Particular pathways of both homologous recombination repair and non-homologous end-joining rely on ERCC1-XPF function. [19] [20] The relevant activity of ERCC1–XPF for both types of double-strand break repair is the ability to remove non-homologous 3′ single-stranded tails from DNA ends before rejoining. This activity is needed during a single-strand annealing subpathway of homologous recombination. Trimming of 3’ single-stranded tail is also needed in a mechanistically distinct subpathway of non-homologous end-joining, dependent on the Ku proteins. [17] Homologous integration of DNA, an important technique for genetic manipulation, is dependent on the function of ERCC1-XPF in the host cell. [21]

Mammalian cells carrying mutations in ERCC1 or XPF are especially sensitive to agents that cause DNA interstrand crosslinks. [22] Interstrand crosslinks block the progression of DNA replication, and structures at blocked DNA replication forks provide substrates for cleavage by ERCC1-XPF. [23] [24] Incisions may be made on either side of the crosslink on one DNA strand to unhook the crosslink and initiate repair. Alternatively, a double-strand break may be made in the DNA near the ICL, and subsequent homologous recombination repair may involve ERCC1-XPF action. Although not the only nuclease involved, ERCC1–XPF is required for ICL repair during several phases of the cell cycle. [25] [26]

Clinical significance

Cerebro-oculo-facio-skeletal syndrome

A few patients with severely disabling ERCC1 mutations that cause cerebro-oculo-facio-skeletal syndrome (COFS) have been reported. [8] [27] COFS syndrome is a rare recessive disorder in which affected individuals undergo rapid neurologic decline and indications of accelerated aging. A very severe case of such disabling mutations is F231L mutation in the tandem helix-hairpin-helix domain of ERCC1 at its interface with XPF. [27] [28] It is shown that this single mutation is very important for the stability of the ERCC1-XPF complex. This Phenylalanine residue is assisting ERCC1 to accommodate a key Phenylalanine residue from XPF (F894) and the mutation (F231L) disturbs this accommodating function. As a consequence, F894 protrudes out of the interface and the mutant complex is dissociating faster compared to the native one. [28] The life span of patients with such mutations is often around 1–2 years. [27]

Cockayne syndrome

One Cockayne syndrome (CS) type II patient designated CS20LO exhibited a homozygous mutation in exon 7 of ERCC1, producing a F231L mutation. [29]

Relevance in chemotherapy

Measuring ERCC1 activity may have utility in clinical cancer medicine because one mechanism of resistance to platinum chemotherapy drugs correlates with high ERCC1 activity. Nucleotide excision repair (NER) is the primary DNA repair mechanism that removes the therapeutic platinum-DNA adducts from the tumor DNA. ERCC1 activity levels, being an important part of the NER common final pathway, may serve as a marker of general NER throughput. This has been suggested for patients with gastric, [30] ovarian and bladder cancers. [31] In Non-small cell lung carcinoma (NSCLC), surgically removed tumors that receive no further therapy have a better survival if ERCC1-positive than if ERCC1-negative. Thus, ERCC1 positivity is a favorable prognostic marker, referring to how the disease will proceed if not further treated. ERCC1-positive NSCLC tumors do not benefit from adjuvant platinum chemotherapy. However, ERCC1-negative NSCLC tumors, prognostically worse without treatment, derive substantial benefit from adjuvant cisplatin-based chemotherapy. High ERCC1 is thus a negative predictive marker, referring to how it will respond to a specific type of treatment. [32] [33] In colorectal cancer, clinical trials have not demonstrated the predictive ability of ERCC1 in oxaliplatin‐based treatment. Thus, European Society for Medical Oncology (ESMO) has not recommended ERCC1 testing prior to the use of oxaliplatin in routine practice. [34] [35] ERCC1 genotyping in humans has shown significant polymorphism at codon 118. [36] These polymorphisms may have differential effects on platinum and mitomycin damage. [36]

Deficiency in cancer

ERCC1 protein expression is reduced or absent in 84% to 100% of colorectal cancers, [37] [38] and lower expression of ERCC1 has been reported as being associated with unfavorable prognosis in patients undergoing treatment with oxaliplatin. [34] The promoter of ERCC1 is methylated in 38% of gliomas, resulting in reduced mRNA and protein expression. [39] The promoter of ERCC1 was located in the DNA 5 kilobases upstream of the protein coding region. [39] Frequencies of epigenetic reductions of nine other DNA repair genes have been evaluated in various cancers and range from 2% ( OGG1 in papillary thyroid cancer) to 88% and 90% ( MGMT in gastric and colon cancers, respectively). Thus, reduction of protein expression of ERCC1 in 84% to 100% of colon cancers indicates that reduced ERCC1 is one of the most frequent reductions of a DNA repair gene observed in a cancer.[ citation needed ] Deficiency in ERCC1 protein expression appears to be an early event in colon carcinogenesis, since ERCC1 was found to be deficient in 40% of the crypts within 10 cm on each side of colonic adenocarcinomas (within the early field defects from which the cancers likely arose). [37]

Cadmium (Cd) and its compounds are well-known human carcinogens. During Cd-induced malignant transformation, the promoter regions of ERCC1, as well as of hMSH2 , XRCC1 , and hOGG1 , were heavily methylated and both the messenger RNA and proteins of these DNA repair genes were progressively reduced. [40] DNA damage also increased with Cd-induced transformation. [40] Reduction of protein expression of ERCC1 in progression to sporadic cancer is unlikely to be due to mutation. While germ line (familial) mutations in DNA repair genes cause a high risk of cancer (see inherited impairment in DNA repair increases cancer risk), somatic mutations in DNA repair genes, including ERCC1, only occur at low levels in sporadic (non-familial) cancers. [41]

Control of ERCC1 protein level occurred at the translational level. In addition to the wild-type sequence, three splice variants of mRNA ERCC1 exist. [42] ERCC1 mRNA is also found to have either wild-type or three alternative transcription start points. Neither the level of overall mRNA transcription, splice variation nor transcription start point of mRNA correlates with protein level of ERCC1. The rate of ERCC1 protein turnover also does not correlate with ERCC1 protein level. A translational level control of ERCC1, due to a microRNA (miRNA), has been shown during HIV viral infection. A trans-activation response element (TAR) miRNA, coded for by the HIV virus, down-regulates ERCC1 protein expression. [43] TAR miRNA allows ERCC1 mRNA to be transcribed, but acts at the p-body level to prevent translation of ERCC1 protein. (A p-body is a cytoplasmic granule “processing body” that interacts with miRNAs to repress translation or trigger degradation of target RNAs.) In breast cancer cell lines, almost one third (55/167) of miRNA promoters were targets for aberrant methylation (epigenetic repression). [44] In breast cancers themselves, methylation of let-7a-3/let-7b miRNA in particular was found. This indicates that let-7a-3/let-7b can be epigenetically repressed.

Repression of let-7a can cause repression of ERCC1 expression through an intermediary step involving the HMGA2 gene. The let-7a miRNA normally represses the HMGA2 gene, and in normal adult tissues, almost no HMGA2 protein is present. [45] (See also Let-7 microRNA precursor.) Reduction or absence of let-7a miRNA allows high expression of the HMGA2 protein. HMGA proteins are characterized by three DNA-binding domains, called AT-hooks, and an acidic carboxy-terminal tail. HMGA proteins are chromatin architectural transcription factors that both positively and negatively regulate the transcription of a variety of genes. They do not display direct transcriptional activation capacity, but regulate gene expression by changing local DNA conformation. Regulation is achieved by binding to AT-rich regions in the DNA and/or direct interaction with several transcription factors. [46] HMGA2 targets and modifies the chromatin architecture at the ERCC1 gene, reducing its expression. [47] Hypermethylation of the promoter for let-7a miRNA reduces its expression and this allows hyperexpression of HMGA2. Hyperexpression of HMGA2 can then reduce expression of ERCC1.

Thus, there are three mechanisms that may be responsible for the low level of protein expression of ERCC1 in 84% to 100% of sporadic colon cancers. From results in gliomas and in cadmium carcinogenesis, methylation of the ERCC1 promoter may be a factor. One or more miRNAs that repress ERCC1 may be a factor. And epigenetically reduced let-7a miRNA allowing hyperexpression of HMGA2 could also reduce protein expression of ERCC1 in colon cancers. Which epigenetic mechanism occurs most frequently, or whether multiple epigenetic mechanisms reduce ERCC1 protein expression in colon cancers has not been determined.[ citation needed ]

Accelerated aging

DNA repair-deficient Ercc1 mutant mice show numerous features of accelerated aging, and have a limited lifespan. [48] Accelerated aging in the mutant involves various organs. Ercc1 mutant mice are deficient in several DNA repair processes including transcription-coupled DNA repair. This deficiency prevents resumption of RNA synthesis on the template DNA strand subsequent to it receiving a transcription-blocking DNA damage. Such blockages of transcription appear to promote premature aging, particularly in non-proliferating or slowly proliferating organs such as the nervous system, liver and kidney [49] (see DNA damage theory of aging).

When Ercc1 mutant mice were subjected to dietary restriction their response closely resembled the beneficial response to dietary restriction of wild-type mice. Dietary restriction extended the lifespan of the Ercc1 mutant mice from 10 to 35 weeks for males and from 13 to 39 weeks for females. [48] It appears that in Ercc1 mutant mice dietary restriction while delaying aging also attenuates accumulation of genome-wide DNA damage and preserves transcriptional output, likely contributing to improved cell viability. [48]

Spermatogenesis and oogenesis

Both male and female Ercc1-deficient mice are infertile. [50] The DNA repair function of Ercc1 appears to be required in both male and female germ cells at all stages of their maturation. The testes of Ercc1-deficient mice have an increased level of 8-oxoguanine in their DNA, suggesting that Ercc1 may have a role in removing oxidative DNA damages.

Notes

Related Research Articles

<span class="mw-page-title-main">Nuclease</span> Class of enzymes which cleave nucleic acids

In biochemistry, a nuclease is an enzyme capable of cleaving the phosphodiester bonds between nucleotides of nucleic acids. Nucleases variously effect single and double stranded breaks in their target molecules. In living organisms, they are essential machinery for many aspects of DNA repair. Defects in certain nucleases can cause genetic instability or immunodeficiency. Nucleases are also extensively used in molecular cloning.

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

DNA repair is a collection of processes by which a cell identifies and corrects damage to the DNA molecules that encodes its genome. In human cells, both normal metabolic activities and environmental factors such as radiation can cause DNA damage, resulting in tens of thousands of individual molecular lesions per cell per day. Many of these lesions cause structural damage to the DNA molecule and can alter or eliminate the cell's ability to transcribe the gene that the affected DNA encodes. Other lesions induce potentially harmful mutations in the cell's genome, which affect the survival of its daughter cells after it undergoes mitosis. As a consequence, the DNA repair process is constantly active as it responds to damage in the DNA structure. When normal repair processes fail, and when cellular apoptosis does not occur, irreparable DNA damage may occur, including double-strand breaks and DNA crosslinkages. This can eventually lead to malignant tumors, or cancer as per the two-hit hypothesis.

<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">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">Base excision repair</span> DNA repair process

Base excision repair (BER) is a cellular mechanism, studied in the fields of biochemistry and genetics, that repairs damaged DNA throughout the cell cycle. It is responsible primarily for removing small, non-helix-distorting base lesions from the genome. The related nucleotide excision repair pathway repairs bulky helix-distorting lesions. BER is important for removing damaged bases that could otherwise cause mutations by mispairing or lead to breaks in DNA during replication. BER is initiated by DNA glycosylases, which recognize and remove specific damaged or inappropriate bases, forming AP sites. These are then cleaved by an AP endonuclease. The resulting single-strand break can then be processed by either short-patch or long-patch BER.

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

MUTYH is a human gene that encodes a DNA glycosylase, MUTYH glycosylase. It is involved in oxidative DNA damage repair and is part of the base excision repair pathway. The enzyme excises adenine bases from the DNA backbone at sites where adenine is inappropriately paired with guanine, cytosine, or 8-oxo-7,8-dihydroguanine, a common form of oxidative DNA damage.

<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">Crosslinking of DNA</span> Phenomenon in genetics

In genetics, crosslinking of DNA occurs when various exogenous or endogenous agents react with two nucleotides of DNA, forming a covalent linkage between them. This crosslink can occur within the same strand (intrastrand) or between opposite strands of double-stranded DNA (interstrand). These adducts interfere with cellular metabolism, such as DNA replication and transcription, triggering cell death. These crosslinks can, however, be repaired through excision or recombination pathways.

<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 H (TFIIH) 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 TFIIH in 1992.

<span class="mw-page-title-main">HMGA2</span> Protein-coding gene in the species Homo sapiens

High-mobility group AT-hook 2, also known as HMGA2, is a protein that, in humans, is encoded by the HMGA2 gene.

<span class="mw-page-title-main">PMS2</span> Protein-coding gene in the species Homo sapiens

Mismatch repair endonuclease PMS2 is an enzyme that in humans is encoded by the PMS2 gene.

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

Xeroderma pigmentosum, complementation group C, also known as XPC, is a protein which in humans is encoded by the XPC gene. XPC is involved in the recognition of bulky DNA adducts in nucleotide excision repair. It is located on chromosome 3.

<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">ERCC6</span> Gene of the species Homo sapiens

DNA excision repair protein ERCC-6 is a protein that in humans is encoded by the ERCC6 gene. The ERCC6 gene is located on the long arm of chromosome 10 at position 11.23.

<span class="mw-page-title-main">ERCC5</span> Protein-coding gene in the species Homo sapiens

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">DCLRE1A</span> Protein-coding gene in the species Homo sapiens

DNA cross-link repair 1A protein is a protein that in humans is encoded by the DCLRE1A gene.

<span class="mw-page-title-main">Orlando D. Schärer</span> Swiss chemist and biologist

Orlando David Schärer is a Swiss chemist and biologist researching DNA repair, genomic integrity, and cancer biology. Schärer has taught biology, chemistry and pharmacology at various university levels on three continents. He is a distinguished professor at the Ulsan National Institute of Science and Technology (UNIST) and an associate director of the IBS Center for Genomic Integrity located in Ulsan, South Korea. He leads the three interdisciplinary research teams in the Chemical & Cancer Biology Branch of the center and specifically heads the Cancer Therapeutics Mechanisms Section.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000012061 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000003549 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Westerveld A, Hoeijmakers JH, van Duin M, de Wit J, Odijk H, Pastink A, et al. (Sep 1984). "Molecular cloning of a human DNA repair gene". Nature. 310 (5976): 425–9. Bibcode:1984Natur.310..425W. doi:10.1038/310425a0. PMID   6462228. S2CID   4336902.
  6. Friedberg EC, Walker GC, Siede W, Wood RD, Schultz RA, Ellenberger T, eds. (2006). DNA Repair and Mutagenesis. ASM Press. p. 286. ISBN   978-1555813192.
  7. "Entrez Gene: ERCC4 excision repair cross-complementing rodent repair deficiency, complementation group 4".
  8. 1 2 Gregg SQ, Robinson AR, Niedernhofer LJ (July 2011). "Physiological consequences of defects in ERCC1-XPF DNA repair endonuclease". DNA Repair. 10 (7): 781–91. doi:10.1016/j.dnarep.2011.04.026. PMC   3139823 . PMID   21612988.
  9. 1 2 Westerveld A, Hoeijmakers JH, van Duin M, de Wit J, Odijk H, Pastink A, et al. (1984). "Molecular cloning of a human DNA repair gene". Nature. 310 (5976): 425–9. Bibcode:1984Natur.310..425W. doi:10.1038/310425a0. PMID   6462228. S2CID   4336902.
  10. Busch D, Greiner C, Lewis K, Ford R, Adair G, Thompson L (September 1989). "Summary of complementation groups of UV-sensitive CHO cell mutants isolated by large-scale screening". Mutagenesis. 4 (5): 349–54. doi:10.1093/mutage/4.5.349. PMID   2687628.
  11. Sgouros J, Gaillard PH, Wood RD (March 1999). "A relationship between a DNA-repair/recombination nuclease family and archaeal helicases". Trends in Biochemical Sciences. 24 (3): 95–7. doi:10.1016/s0968-0004(99)01355-9. PMID   10203755.
  12. Enzlin JH, Schärer OD (April 2002). "The active site of the DNA repair endonuclease XPF-ERCC1 forms a highly conserved nuclease motif". The EMBO Journal. 21 (8): 2045–53. doi:10.1093/emboj/21.8.2045. PMC   125967 . PMID   11953324.
  13. Gaillard PH, Wood RD (February 2001). "Activity of individual ERCC1 and XPF subunits in DNA nucleotide excision repair". Nucleic Acids Research. 29 (4): 872–9. doi:10.1093/nar/29.4.872. PMC   29621 . PMID   11160918.
  14. 1 2 Sijbers AM, de Laat WL, Ariza RR, Biggerstaff M, Wei YF, Moggs JG, et al. (September 1996). "Xeroderma pigmentosum group F caused by a defect in a structure-specific DNA repair endonuclease" (PDF). Cell. 86 (5): 811–22. doi:10.1016/s0092-8674(00)80155-5. hdl: 1765/3110 . PMID   8797827. S2CID   12957716.
  15. Bardwell AJ, Bardwell L, Tomkinson AE, Friedberg EC (September 1994). "Specific cleavage of model recombination and repair intermediates by the yeast Rad1-Rad10 DNA endonuclease". Science. 265 (5181): 2082–5. Bibcode:1994Sci...265.2082B. doi:10.1126/science.8091230. PMID   8091230.
  16. 1 2 McNeil EM, Melton DW (November 2012). "DNA repair endonuclease ERCC1-XPF as a novel therapeutic target to overcome chemoresistance in cancer therapy". Nucleic Acids Research. 40 (20): 9990–10004. doi:10.1093/nar/gks818. PMC   3488251 . PMID   22941649.
  17. 1 2 Ahmad A, Robinson AR, Duensing A, van Drunen E, Beverloo HB, Weisberg DB, et al. (August 2008). "ERCC1-XPF endonuclease facilitates DNA double-strand break repair". Molecular and Cellular Biology. 28 (16): 5082–92. doi:10.1128/MCB.00293-08. PMC   2519706 . PMID   18541667.
  18. Wood RD, Burki HJ, Hughes M, Poley A (February 1983). "Radiation-induced lethality and mutation in a repair-deficient CHO cell line". International Journal of Radiation Biology and Related Studies in Physics, Chemistry and Medicine. 43 (2): 207–13. doi:10.1080/09553008314550241. PMID   6600735.
  19. Al-Minawi AZ, Saleh-Gohari N, Helleday T (January 2008). "The ERCC1/XPF endonuclease is required for efficient single-strand annealing and gene conversion in mammalian cells". Nucleic Acids Research. 36 (1): 1–9. doi:10.1093/nar/gkm888. PMC   2248766 . PMID   17962301.
  20. Sargent RG, Rolig RL, Kilburn AE, Adair GM, Wilson JH, Nairn RS (November 1997). "Recombination-dependent deletion formation in mammalian cells deficient in the nucleotide excision repair gene ERCC1". Proceedings of the National Academy of Sciences of the United States of America. 94 (24): 13122–7. Bibcode:1997PNAS...9413122S. doi: 10.1073/pnas.94.24.13122 . PMC   24273 . PMID   9371810.
  21. Niedernhofer LJ, Essers J, Weeda G, Beverloo B, de Wit J, Muijtjens M, et al. (November 2001). "The structure-specific endonuclease Ercc1-Xpf is required for targeted gene replacement in embryonic stem cells". The EMBO Journal. 20 (22): 6540–9. doi:10.1093/emboj/20.22.6540. PMC   125716 . PMID   11707424.
  22. Wood RD (July 2010). "Mammalian nucleotide excision repair proteins and interstrand crosslink repair". Environmental and Molecular Mutagenesis. 51 (6): 520–6. Bibcode:2010EnvMM..51..520W. doi:10.1002/em.20569. PMC   3017513 . PMID   20658645.
  23. Klein Douwel D, Boonen RA, Long DT, Szypowska AA, Räschle M, Walter JC, Knipscheer P (May 2014). "XPF-ERCC1 acts in Unhooking DNA interstrand crosslinks in cooperation with FANCD2 and FANCP/SLX4". Molecular Cell. 54 (3): 460–71. doi:10.1016/j.molcel.2014.03.015. PMC   5067070 . PMID   24726325.
  24. Kuraoka I, Kobertz WR, Ariza RR, Biggerstaff M, Essigmann JM, Wood RD (August 2000). "Repair of an interstrand DNA cross-link initiated by ERCC1-XPF repair/recombination nuclease". The Journal of Biological Chemistry. 275 (34): 26632–6. doi: 10.1074/jbc.C000337200 . PMID   10882712.
  25. Clauson C, Schärer OD, Niedernhofer L (October 2013). "Advances in understanding the complex mechanisms of DNA interstrand cross-link repair". Cold Spring Harbor Perspectives in Biology. 5 (10): a012732. doi:10.1101/cshperspect.a012732. PMC   4123742 . PMID   24086043.
  26. Rahn JJ, Adair GM, Nairn RS (July 2010). "Multiple roles of ERCC1-XPF in mammalian interstrand crosslink repair". Environmental and Molecular Mutagenesis. 51 (6): 567–81. Bibcode:2010EnvMM..51..567R. doi:10.1002/em.20583. PMID   20658648. S2CID   29240680.
  27. 1 2 3 Jaspers NG, Raams A, Silengo MC, Wijgers N, Niedernhofer LJ, Robinson AR, et al. (March 2007). "First reported patient with human ERCC1 deficiency has cerebro-oculo-facio-skeletal syndrome with a mild defect in nucleotide excision repair and severe developmental failure". American Journal of Human Genetics. 80 (3): 457–66. doi:10.1086/512486. PMC   1821117 . PMID   17273966.
  28. 1 2 Faridounnia M, Wienk H, Kovačič L, Folkers GE, Jaspers NG, Kaptein R, et al. (August 2015). "The Cerebro-oculo-facio-skeletal Syndrome Point Mutation F231L in the ERCC1 DNA Repair Protein Causes Dissociation of the ERCC1-XPF Complex". The Journal of Biological Chemistry. 290 (33): 20541–55. doi: 10.1074/jbc.M114.635169 . PMC   4536458 . PMID   26085086.
  29. Kashiyama K, Nakazawa Y, Pilz DT, Guo C, Shimada M, Sasaki K, et al. (May 2013). "Malfunction of nuclease ERCC1-XPF results in diverse clinical manifestations and causes Cockayne syndrome, xeroderma pigmentosum, and Fanconi anemia". American Journal of Human Genetics. 92 (5): 807–19. doi:10.1016/j.ajhg.2013.04.007. PMC   3644632 . PMID   23623389.
  30. Kwon HC, Roh MS, Oh SY, Kim SH, Kim MC, Kim JS, Kim HJ (March 2007). "Prognostic value of expression of ERCC1, thymidylate synthase, and glutathione S-transferase P1 for 5-fluorouracil/oxaliplatin chemotherapy in advanced gastric cancer". Annals of Oncology. 18 (3): 504–9. doi: 10.1093/annonc/mdl430 . PMID   17322540.
  31. Bellmunt J, Paz-Ares L, Cuello M, Cecere FL, Albiol S, Guillem V, et al. (March 2007). "Gene expression of ERCC1 as a novel prognostic marker in advanced bladder cancer patients receiving cisplatin-based chemotherapy". Annals of Oncology. 18 (3): 522–8. doi: 10.1093/annonc/mdl435 . PMID   17229776.
  32. Olaussen KA, Dunant A, Fouret P, Brambilla E, André F, Haddad V, et al. (September 2006). "DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy". The New England Journal of Medicine. 355 (10): 983–91. doi: 10.1056/NEJMoa060570 . PMID   16957145.
  33. Soria JC (July 2007). "ERCC1-tailored chemotherapy in lung cancer: the first prospective randomized trial". Journal of Clinical Oncology. 25 (19): 2648–9. doi:10.1200/JCO.2007.11.3167. PMID   17602070.
  34. 1 2 Yau TO (October 2019). "Precision treatment in colorectal cancer: Now and the future". JGH Open. 3 (5): 361–369. doi:10.1002/jgh3.12153. PMC   6788378 . PMID   31633039.
  35. Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. (August 2016). "ESMO consensus guidelines for the management of patients with metastatic colorectal cancer". Annals of Oncology. 27 (8): 1386–422. doi: 10.1093/annonc/mdw235 . hdl: 10400.26/14245 . PMID   27380959.
  36. 1 2 Bohanes P, Labonte MJ, Lenz HJ (September 2011). "A review of excision repair cross-complementation group 1 in colorectal cancer". Clinical Colorectal Cancer. 10 (3): 157–64. doi:10.1016/j.clcc.2011.03.024. PMID   21855036.
  37. 1 2 Facista A, Nguyen H, Lewis C, Prasad AR, Ramsey L, Zaitlin B, et al. (April 2012). "Deficient expression of DNA repair enzymes in early progression to sporadic colon cancer". Genome Integrity. 3 (1): 3. doi: 10.1186/2041-9414-3-3 . PMC   3351028 . PMID   22494821.
  38. Smith DH, Fiehn AM, Fogh L, Christensen IJ, Hansen TP, Stenvang J, et al. (March 2014). "Measuring ERCC1 protein expression in cancer specimens: validation of a novel antibody". Scientific Reports. 4: 4313. Bibcode:2014NatSR...4E4313S. doi:10.1038/srep04313. PMC   3945488 . PMID   24603753.
  39. 1 2 Chen HY, Shao CJ, Chen FR, Kwan AL, Chen ZP (April 2010). "Role of ERCC1 promoter hypermethylation in drug resistance to cisplatin in human gliomas". International Journal of Cancer. 126 (8): 1944–1954. doi: 10.1002/ijc.24772 . PMID   19626585. S2CID   3423262.
  40. 1 2 Zhou ZH, Lei YX, Wang CX (February 2012). "Analysis of aberrant methylation in DNA repair genes during malignant transformation of human bronchial epithelial cells induced by cadmium". Toxicological Sciences. 125 (2): 412–7. doi: 10.1093/toxsci/kfr320 . PMID   22112500.
  41. Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, et al. (November 2007). "The genomic landscapes of human breast and colorectal cancers". Science. 318 (5853): 1108–13. Bibcode:2007Sci...318.1108W. CiteSeerX   10.1.1.218.5477 . doi:10.1126/science.1145720. PMID   17932254. S2CID   7586573.
  42. McGurk CJ, Cummings M, Köberle B, Hartley JA, Oliver RT, Masters JR (April 2006). "Regulation of DNA repair gene expression in human cancer cell lines". Journal of Cellular Biochemistry. 97 (5): 1121–36. doi: 10.1002/jcb.20711 . PMID   16315315. S2CID   24969413.
  43. Klase Z, Winograd R, Davis J, Carpio L, Hildreth R, Heydarian M, et al. (February 2009). "HIV-1 TAR miRNA protects against apoptosis by altering cellular gene expression". Retrovirology. 6: 18. doi: 10.1186/1742-4690-6-18 . PMC   2654423 . PMID   19220914.
  44. Vrba L, Muñoz-Rodríguez JL, Stampfer MR, Futscher BW (2013). "miRNA gene promoters are frequent targets of aberrant DNA methylation in human breast cancer". PLOS ONE. 8 (1): e54398. Bibcode:2013PLoSO...854398V. doi: 10.1371/journal.pone.0054398 . PMC   3547033 . PMID   23342147.
  45. Motoyama K, Inoue H, Nakamura Y, Uetake H, Sugihara K, Mori M (April 2008). "Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family". Clinical Cancer Research. 14 (8): 2334–40. doi: 10.1158/1078-0432.CCR-07-4667 . PMID   18413822.
  46. Cleynen I, Van de Ven WJ (February 2008). "The HMGA proteins: a myriad of functions (Review)". International Journal of Oncology. 32 (2): 289–305. doi: 10.3892/ijo.32.2.289 . PMID   18202751.
  47. Borrmann L, Schwanbeck R, Heyduk T, Seebeck B, Rogalla P, Bullerdiek J, Wisniewski JR (December 2003). "High mobility group A2 protein and its derivatives bind a specific region of the promoter of DNA repair gene ERCC1 and modulate its activity". Nucleic Acids Research. 31 (23): 6841–51. doi:10.1093/nar/gkg884. PMC   290254 . PMID   14627817.
  48. 1 2 3 Vermeij WP, Dollé ME, Reiling E, Jaarsma D, Payan-Gomez C, Bombardieri CR, et al. (September 2016). "Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice". Nature. 537 (7620): 427–431. Bibcode:2016Natur.537..427V. doi:10.1038/nature19329. PMC   5161687 . PMID   27556946.
  49. Marteijn JA, Lans H, Vermeulen W, Hoeijmakers JH (July 2014). "Understanding nucleotide excision repair and its roles in cancer and ageing". Nature Reviews. Molecular Cell Biology. 15 (7): 465–81. doi:10.1038/nrm3822. PMID   24954209. S2CID   9174323.
  50. Hsia KT, Millar MR, King S, Selfridge J, Redhead NJ, Melton DW, Saunders PT (January 2003). "DNA repair gene Ercc1 is essential for normal spermatogenesis and oogenesis and for functional integrity of germ cell DNA in the mouse". Development. 130 (2): 369–78. doi:10.1242/dev.00221. PMID   12466203. S2CID   15898657.

Further reading