Download Gene Section RRM1 (ribonucleotide reductase M1) Atlas of Genetics and Cytogenetics

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Atlas of Genetics and Cytogenetics
in Oncology and Haematology
OPEN ACCESS JOURNAL AT INIST-CNRS
Gene Section
Review
RRM1 (ribonucleotide reductase M1)
Scott N Freeman, Gerold Bepler
Department of Thoracic Oncology, Experimental Therapeutics Program and Lung Cancer Program, H Lee
Moffitt Cancer Center and Research Institute, Tampa, Florida, USA (SNF, GB)
Published in Atlas Database: June 2009
Online updated version : http://AtlasGeneticsOncology.org/Genes/RRM1ID42174ch11p15.html
DOI: 10.4267/2042/44758
This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence.
© 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology
Description
Identity
The RRM1 protein is 792 amino acids in length and has
a calculated molecular weight of 90 kDa. Analysis of
the human RRM1 protein via the NCBI Conserved
Domains program reveals an N-terminal ATP-cone
domain and a Ribonucleotide Reductase (RNR) Class I
domain which spans the majority of the protein. The
RNR Class I domain harbors the all-alpha domain, the
barrel domain, as well as regions and residues
comprising the active site, the allosteric effector dTTPbinding site, the RRM2 peptide-binding site and the
dimer interface.
Other names: R1; RIR1; RR1
HGNC (Hugo): RRM1
Location: 11p15.4
Local order: STIM1 - RRM1 - OR55B1P LOC643244.
Note: The RRM1 gene is oriented on the plus strand.
The chromosomal region containing the RRM1 gene
locus displays frequent loss of heterozygosity (LOH) in
lung cancer and is amplified in some instances of
acquired resistance to gemcitabine and hydroxyurea.
Expression
DNA/RNA
Pseudogene
While the RRM2 subunit of the ribonucleotide
reductase holoenzyme is regulated in a cell cyclespecific manner, RRM1 expression remains relatively
constant in actively proliferating cells. However,
expression of RRM1 is significantly decreased upon
exit from the cell cycle to G0 or terminal
differentiation. This decreased level of RRM1
expression in non-dividing cells is essential for the
supply of dNTPs for mitochondrial DNA replication.
Upregulation of RRM1 occurs when cells are
stimulated to re-enter the cell cycle from G0.
There are no reported human RRM1 pseudogenes.
Localisation
Protein
Depending on the experimental technique employed,
the cell line or tissue sample, the cellular state and the
antibody source, RRM1 can be detected within the
nucleus, the cytoplasm or both. In quantitative
immunofluorescent staining of non-small-cell lung
cancer (NSCLC) cell lines and tumor samples, RRM1
protein is predominantly nuclear. The method of
analyzing nuclear RRM1 expression in resected
NSCLC tumor samples can be used for survival
prognostication. This is in agreement with fractionation
of NSCLC cell lines, where RRM1 is detected almost
Description
The RRM1 gene consists of 44182 base pairs which
encode 19 exons.
Transcription
The mature RRM1 mRNA is 3234 ribonucleotides in
length and harbors an open reading frame of 2379
ribonucleotides.
Note
The RRM1 protein functions in a heterodimeric
tetramer with either RRM2 (RRM2a) or p53R2
(RRM2b) in the ribonucleotide reductase holoenzyme.
While this interaction is essential for the de novo
synthesis of deoxyribonucleotides, it is unclear whether
this association is necessary for other biological
activities ascribed to RRM1.
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
473
RRM1 (ribonucleotide reductase M1)
Freeman SN, Bepler G
promotes the efficient repair of DNA damage.
Additionally, RRM1-overexpressing cells have an
increased level of apoptosis. When subjected to a
murine chemical carcinogenesis protocol, mice
transgenic for RRM1 (tg+) have significantly reduced
lung tumor formation, and splenocytes derived from
tg+ mice repair hydrogen peroxide-induced DNA
damage more efficiently than those from tg- littermates.
While the exact mechanism by which RRM1
contributes to DNA damage response and repair in
unclear, it may involve its association with the p53R2
RR subunit. This subunit is induced by the DNA
damage responsive p53 and p73 transcription factors, is
stabilized by ATM-mediated phosphorylation and can
substitute for RRM2 in the RR complex. Current data
indicates a role for p53R2 in providing dNTPs during
DNA damage repair and may explain the involvement
of RRM1 in DNA damage response and repair.
With regard to tumor suppression, overexpression of
RRM1 in human and mouse lung cancer cell lines, as
well as ras-transformed mouse fibroblasts, inhibits
metastasis, tumorigenicity and motility. It also
suppresses metastasis formation and increases survival
in a syngeneic murine lung cancer model. Evidence
points to reduced phosphorylation of focal adhesion
kinase (FAK) and increased PTEN expression as
mediators of these effects. Mice transgenic for RRM1
subjected to a murine chemical carcinogenesis protocol
display
significantly
decreased
lung
tumor
development and survive significantly longer than tglittermates. Additionally, multiple clinical studies show
correlations between increased intratumoral RRM1
expression in surgically resected tumor tissue and
increased survival (NSCLC, pancreatic cancer, bladder
cancer). As previously noted, reduced FAK
phosphorylation and increased PTEN expression are
thought to be the primary mediators of the tumor
suppressor function of RRM1; however, the ability of
RRM1 to contribute to DNA damage response and
repair may also play a role.
exclusively in the nuclear fraction. In contrast, studies
utilizing various experimental methods to examine
RRM1 in mouse and human lung fibroblasts find
RRM1 to be cytoplasmic. This is in line with other
studies that find RRM1 localized in cytoplasmic
compartments in mouse fibroblasts, as well as
cytoplasmic RRM1 immunoreactivity in various rat
tissues. Still, another study demonstrates both nuclear
and cytoplasmic RRM1 in multiple cell lines and finds
RRM1 localized to the nuclear membrane, while others
report a DNA damage-dependent relocalization of
RRM1 from the cytoplasm to the nucleus. Given the
current evidence, it is likely that RRM1 localization is
dependent on cell type, tissue of origin, and cellular
state. It is also evident that variables such as the
antibody and experimental technique employed obscure
a conclusive answer as to the true localization
characteristics of RRM1.
Function
The
RRM1
protein
is
implicated
in
deoxyribonucleotide (dNTP) synthesis, DNA damage
response and repair and tumor suppression. In dNTP
synthesis, RRM1 serves as the regulatory subunit of the
ribonucleotide reductase (RR) holoenzyme, which
catalyzes the de novo synthesis of dNTPs from
ribonucleotide precursors. Synthesis of dNTPs via this
pathway is reported to occur in the cytosol, with dNTPs
re-localizing to the nucleus through diffusion and to the
mitochondria via transport mechanisms. The RR
enzyme primarily exists as a heterodimeric tetramer of
RRM1 and RRM2, the primary catalytic subunit. While
RRM1 expression remains relatively constant in
cycling cells, RRM2 is regulated in a cell cycle-specific
manner and is degraded upon exit from mitosis and
absent in quiescent cells. Given this, the RRM2 subunit
is the primary regulator of RR enzymatic activity.
RRM1 can alternatively associate with the p53R2
subunit in substitution for RRM2. Unlike RRM2,
p53R2 is present in quiescent cells and as such
associates with RRM1 under this context to provide
dNTPs for mitochondrial DNA (mtDNA) replication in
cells which are not actively dividing. Additionally, the
expression of p53R2 is induced by the DNA damage
response proteins p53 and p73, stabilized by ATMmediated phosphorylation and is reported to be
involved in the supply of dNTPs by RR for mtDNA
replication and DNA damage repair. The role of RRM1
in the synthesis of dNTPs is also manifested as
upregulation in cases of acquired resistance to
gemcitabine and hydroxyurea, which directly inhibit
the RR holoenzyme.
In the context of DNA damage response and repair,
stable
overexpression
of
RRM1
in
lung
adenocarcinoma cells results in an increased fraction of
cells arrested in G2/M. This coincides with increased
expression of the GADD45 G2 checkpoint protein,
which is thought to mediate this arrest. In response to
DNA-damaging agents, overexpression of RRM1
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
Homology
The RRM1 protein is highly conserved from human to
many lower organisms. Human RRM1 protein shares
99.7% similarity and 97.6% identity with mouse (M.
musculus) RRM1 and 88.2% similarity and 69.1%
identity with that of fission yeast (S. pombe). It does
not harbor significant homology to other human
proteins.
Mutations
Germinal
At this point, there are no described germline mutations
within the RRM1 gene. There are a total of 272
reported single nucleotide polymorphisms (SNPs) for
the RRM1 gene. Nine are in the coding region of
RRM1, and four result in amino acid alterations which
consist of: G249A, A768C, T821G and T2565C. We
474
RRM1 (ribonucleotide reductase M1)
Freeman SN, Bepler G
and decreased overall survival (OS) in patients with
stage I or stage II disease. RRM1 mRNA derived from
surgically resected NSCLC tumors indicates that high
levels of RRM1 expression are associated with longer
survival. This result was confirmed in a study where an
elevated level of intratumoral RRM1 protein was
prognostic of favorable survival. In addition to RRM1
expression, promoter polymorphisms at positions -37
and -524 (RR37AC and RR524CT), which regulate
RRM1 expression in in vitro models, are associated
with survival.
Cytogenetics
The RRM1 gene resides at 11p15.5, a region frequently
associated with allele loss in NSCLC. This is
exemplified in primary lung cancer tissue as well as
lung cancer cell lines. The region of minimal allele loss
in NSCLC is mapped to a 310 kb region which contains
the complete coding sequence for the RRM1 and
SSA/Ro52 genes. Of these two genes, RRM1 is
identified as the putative tumor suppressor within this
region of LOH.
sequenced the genomic region of RRM1 and deposited
this data in GenBank (AF107045), which provided for
the reference for some of the reported SNPs. In
addition, we described SNPs in the RRM1 promoter
region that have a substantial effect on in vitro reporter
gene transcription.
Somatic
RRM1 resides at 11p15.5, a frequent region of allele
loss in cancer. Additionally, amplification of RRM1
has been observed in acquired resistance to
gemcitabine and hydroxyurea.
Implicated in
Non-small cell lung cancer (NSCLC)
Note
Overexpression of RRM1 in human and mouse lung
cancer cell lines inhibits motility in cell culture and
decreases metastasis and tumorigenicity in a syngeneic
mouse lung cancer model. Increased survival is also
noted upon RRM1 overexpression in the same model.
Mice transgenic for RRM1 subjected to a chemical
carcinogenesis protocol display significantly decreased
lung tumor development and survive significantly
longer than tg- littermates.
Since gemcitabine works in part through the inhibition
of ribonucleotide reductase, RRM1 is a key molecule
involved in the therapeutic response. Multiple NSCLC
clinical studies demonstrate that while decreased
intratumoral RRM1 is indicative of sensitivity to
gemcitabine, elevated levels are predictive of poor
response to gemcitabine. In NSCLC cell lines,
depletion of RRM1 results in sensitivity to
gemcitabine, and vice versa increased RRM1
expression results in resistance to gemcitabine.
Gemcitabine-resistant
NSCLC
cells
display
upregulation of RRM1, and depletion of RRM1 in
these
cells
restores
gemcitabine
sensitivity.
Additionally, a 2464G>A RRM1 polymorphism was
reported to be associated with gemcitabine sensitivity
in cancer cell lines of various origins, while NSCLC
patients harboring a combination of the RR37AC and
RR524CT promoter polymorphisms, the haplotype
associated with decreased promoter activity, show
greater response to gemcitabine.
Pancreatic cancer
Note
Increased RRM1 expression is indicative of resistance
to gemcitabine in pancreatic cancer. RRM1 is the most
highly upregulated gene observed in the 81-fold
gemcitabine-resistant MiaPaCa2 derived cell line
MiaPaCa2-RG, and siRNA depletion of RRM1
expression in this resistant cell line restores
gemcitabine sensitivity. During the acquisition of
gemcitabine resistance in pancreatic cell lines,
progressive upregulation of RRM1 is observed.
Disease
The term pancreatic cancer refers to malignant
neoplasms of the pancreas. Tumors of the pancreas can
originate from either exocrine or endocrine pancreatic
cells, with exocrine pancreatic cancer being the
predominant form. The majority of exocrine pancreatic
cancers are adenocarcinomas. The NCI estimates that
there will be 42,470 new cases of pancreatic cancer and
35,240 deaths in 2009.
Biliary tract cancer
Note
Depletion of RRM1 expression via siRNA in the G-415
biliary tract carcinoma cell line increases sensitivity to
gemcitabine and enhances gemcitabine-induced
apoptosis. Tumor tissue derived from biliary tract
carcinoma patients treated with gemcitabine shows a
tendency toward elevated RRM1 expression
associating with progressive disease (PD) and
decreased RRM1 expression associating with partial
response (PR), although these tendencies were not
statistically significant. These results suggest that
RRM1 expression may be a useful tool for predicting
response to gemcitabine in patients with cancer of the
biliary tract.
Disease
NSCLC is the most frequently diagnosed type of lung
cancer and refers to malignant neoplasms of the lung
which are histologically distinct from small cell lung
cancer. NSCLC cases are classified as adenocarcinoma,
large cell carcinoma or squamous (epidermoid)
carcinoma. The National Cancer Institute (NCI)
estimates that there were 215,020 new cases of lung
cancer and 161,840 deaths in 2008.
Prognosis
Loss of the chromosomal region harboring RRM1 in
NSCLC is associated with increased metastatic spread
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
475
RRM1 (ribonucleotide reductase M1)
Freeman SN, Bepler G
2',2'-difluorodeoxycytidine
arabinofuranosylcytosine.
15;48(14):4024-31
Disease
Biliary tract cancer refers to malignant neoplasms of
the gallbladder and/or bile duct. Cancers of the bile
duct are referred to as cholangiocarcinomas, while
other cancers of the biliary system include gallbladder
cancers and cancer of the ampulla of Vater. Biliary
tract malignancies are relatively rare.
and
Cancer
Res.
1-beta-D1988
Jul
Mann GJ, Musgrove EA, Fox RM, Thelander L. Ribonucleotide
reductase M1 subunit in cellular proliferation, quiescence, and
differentiation. Cancer Res. 1988 Sep 15;48(18):5151-6
Weston A, Willey JC, Modali R, Sugimura H, McDowell EM,
Resau J, Light B, Haugen A, Mann DL, Trump BF. Differential
DNA sequence deletions from chromosomes 3, 11, 13, and 17
in squamous-cell carcinoma, large-cell carcinoma, and
adenocarcinoma of the human lung. Proc Natl Acad Sci U S A.
1989 Jul;86(13):5099-103
References
Thelander L, Reichard P. Reduction of ribonucleotides. Annu
Rev Biochem. 1979;48:133-58
Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey
GB, Plunkett W. Inhibition of ribonucleotide reduction in CCRFCEM cells by 2',2'-difluorodeoxycytidine. Mol Pharmacol. 1990
Oct;38(4):567-72
Thelander L, Eriksson S, Akerman M. Ribonucleotide
reductase from calf thymus. Separation of the enzyme into two
nonidentical subunits, proteins M1 and M2. J Biol Chem. 1980
Aug 10;255(15):7426-32
Engström Y, Rozell B, Hansson HA, Stemme S, Thelander L.
Localization of ribonucleotide reductase in mammalian cells.
EMBO J. 1984 Apr;3(4):863-7
Hurta RA, Wright JA. Amplification of the genes for both
components of ribonucleotide reductase in hydroxyurea
resistant mammalian cells. Biochem Biophys Res Commun.
1990 Feb 28;167(1):258-64
Engström Y, Eriksson S, Jildevik I, Skog S, Thelander L,
Tribukait B. Cell cycle-dependent expression of mammalian
ribonucleotide reductase. Differential regulation of the two
subunits. J Biol Chem. 1985 Aug 5;260(16):9114-6
Sikorska M, Brewer LM, Youdale T, Richards R, Whitfield JF,
Houghten RA, Walker PR. Evidence that mammalian
ribonucleotide reductase is a nuclear membrane associated
glycoprotein. Biochem Cell Biol. 1990 May;68(5):880-8
McClarty GA, Chan AK, Wright JA. Characterization of a
mouse cell line selected for hydroxyurea resistance by a
stepwise procedure: drug-dependent overproduction of
ribonucleotide reductase activity. Somat Cell Mol Genet. 1986
Mar;12(2):121-31
Ludwig CU, Raefle G, Dalquen P, Stulz P, Stahel R, Obrecht
JP. Allelic loss on the short arm of chromosome 11 in nonsmall-cell lung cancer. Int J Cancer. 1991 Nov 11;49(5):661-5
Parker NJ, Begley CG, Fox RM. Human M1 subunit of
ribonucleotide reductase: cDNA sequence and expression in
stimulated lymphocytes. Nucleic Acids Res. 1991 Jul
11;19(13):3741
Cocking JM, Tonin PN, Stokoe NM, Wensing EJ, Lewis WH,
Srinivasan PR. Gene for M1 subunit of ribonucleotide
reductase is amplified in hydroxyurea-resistant hamster cells.
Somat Cell Mol Genet. 1987 May;13(3):221-33
Pavloff N, Rivard D, Masson S, Shen SH, Mes-Masson AM.
Sequence analysis of the large and small subunits of human
ribonucleotide reductase. DNA Seq. 1992;2(4):227-34
McClarty GA, Chan AK, Engstrom Y, Wright JA, Thelander L.
Elevated expression of M1 and M2 components and druginduced posttranscriptional modulation of ribonucleotide
reductase in a hydroxyurea-resistant mouse cell line.
Biochemistry. 1987 Dec 1;26(24):8004-11
Bepler G, Garcia-Blanco MA. Three tumor-suppressor regions
on chromosome 11p identified by high-resolution deletion
mapping in human non-small-cell lung cancer. Proc Natl Acad
Sci U S A. 1994 Jun 7;91(12):5513-7
Tonin PN, Stallings RL, Carman MD, Bertino JR, Wright JA,
Srinivasan PR, Lewis WH. Chromosomal assignment of
amplified genes in hydroxyurea-resistant hamster cells.
Cytogenet Cell Genet. 1987;45(2):102-8
Fong KM, Zimmerman PV, Smith PJ. Correlation of loss of
heterozygosity at 11p with tumour progression and survival in
non-small cell lung cancer. Genes Chromosomes Cancer.
1994 Jul;10(3):183-9
Wright JA, Alam TG, McClarty GA, Tagger AY, Thelander L.
Altered expression of ribonucleotide reductase and role of M2
gene amplification in hydroxyurea-resistant hamster, mouse,
rat, and human cell lines. Somat Cell Mol Genet. 1987
Mar;13(2):155-65
Fong KM, Zimmerman PV, Smith PJ. Microsatellite instability
and other molecular abnormalities in non-small cell lung
cancer. Cancer Res. 1995 Jan 1;55(1):28-30
Tran YK, Newsham IF. High-density marker analysis of
11p15.5 in non-small cell lung carcinomas reveals allelic
deletion of one shared and one distinct region when compared
to breast carcinomas. Cancer Res. 1996 Jul 1;56(13):2916-21
Brissenden JE, Caras I, Thelander L, Francke U. The structural
gene for the M1 subunit of ribonucleotide reductase maps to
chromosome 11, band p15, in human and to chromosome 7 in
mouse. Exp Cell Res. 1988 Jan;174(1):302-8
Fan H, Huang A, Villegas C, Wright JA. The R1 component of
mammalian ribonucleotide reductase has malignancysuppressing activity as demonstrated by gene transfer
experiments. Proc Natl Acad Sci U S A. 1997 Nov
25;94(24):13181-6
Choy BK, McClarty GA, Chan AK, Thelander L, Wright JA.
Molecular mechanisms of drug resistance involving
ribonucleotide reductase: hydroxyurea resistance in a series of
clonally related mouse cell lines selected in the presence of
increasing drug concentrations. Cancer Res. 1988 Apr
15;48(8):2029-35
O'Briant K, Jolicoeur E, Garst J, Campa M, Schreiber G,
Bepler G. Growth inhibition of a human lung adenocarcinoma
cell line by genetic complementation with chromosome 11.
Anticancer Res. 1997 Sep-Oct;17(5A):3243-51
Engström Y, Rozell B. Immunocytochemical evidence for the
cytoplasmic localization and differential expression during the
cell cycle of the M1 and M2 subunits of mammalian
ribonucleotide reductase. EMBO J. 1988 Jun;7(6):1615-20
O'Briant KC, Bepler G. Delineation of the centromeric and
telomeric chromosome segment 11p15.5 lung cancer
suppressor regions LOH11A and LOH11B. Genes
Chromosomes Cancer. 1997 Feb;18(2):111-4
Heinemann V, Hertel LW, Grindey GB, Plunkett W.
Comparison of the cellular pharmacokinetics and toxicity of
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
476
RRM1 (ribonucleotide reductase M1)
Freeman SN, Bepler G
Bepler G, Fong KM, Johnson BE, O'Briant KC, Daly LA,
Zimmerman PV, Garcia-Blanco MA, Peterson B. Association of
chromosome 11 locus D11S12 with histology, stage, and
metastases in lung cancer. Cancer Detect Prev.
1998;22(1):14-9
Bepler G, Sharma S, Cantor A, Gautam A, Haura E, Simon G,
Sharma A, Sommers E, Robinson L. RRM1 and PTEN as
prognostic parameters for overall and disease-free survival in
patients with non-small-cell lung cancer. J Clin Oncol. 2004
May 15;22(10):1878-85
Bepler G, O'briant KC, Kim YC, Schreiber G, Pitterle DM. A
1.4-Mb high-resolution physical map and contig of
chromosome segment 11p15.5 and genes in the LOH11A
metastasis suppressor region. Genomics. 1999 Jan
15;55(2):164-75
Davidson JD, Ma L, Flagella M, Geeganage S, Gelbert LM,
Slapak CA. An increase in the expression of ribonucleotide
reductase large subunit 1 is associated with gemcitabine
resistance in non-small cell lung cancer cell lines. Cancer Res.
2004 Jun 1;64(11):3761-6
Bridges EG, Jiang Z, Cheng YC. Characterization of a dCTP
transport activity reconstituted from human mitochondria. J Biol
Chem. 1999 Feb 19;274(8):4620-5
Rosell R, Danenberg KD, Alberola V, Bepler G, Sanchez JJ,
Camps C, Provencio M, Isla D, Taron M, Diz P, Artal A.
Ribonucleotide reductase messenger RNA expression and
survival in gemcitabine/cisplatin-treated advanced non-small
cell lung cancer patients. Clin Cancer Res. 2004 Feb
15;10(4):1318-25
Pitterle DM, Kim YC, Jolicoeur EM, Cao Y, O'Briant KC, Bepler
G. Lung cancer and the human gene for ribonucleotide
reductase subunit M1 (RRM1). Mamm Genome. 1999
Sep;10(9):916-22
Rosell R, Felip E, Taron M, Majo J, Mendez P, Sanchez-Ronco
M, Queralt C, Sanchez JJ, Maestre J. Gene expression as a
predictive marker of outcome in stage IIB-IIIA-IIIB non-small
cell lung cancer after induction gemcitabine-based
chemotherapy followed by resectional surgery. Clin Cancer
Res. 2004 Jun 15;10(12 Pt 2):4215s-4219s
Chabes A, Thelander L. Controlled protein degradation
regulates ribonucleotide reductase activity in proliferating
mammalian cells during the normal cell cycle and in response
to DNA damage and replication blocks. J Biol Chem. 2000 Jun
9;275(23):17747-53
Bepler G, Zheng Z, Gautam A, Sharma S, Cantor A, Sharma
A, Cress WD, Kim YC, Rosell R, McBride C, Robinson L,
Sommers E, Haura E. Ribonucleotide reductase M1 gene
promoter activity, polymorphisms, population frequencies, and
clinical relevance. Lung Cancer. 2005 Feb;47(2):183-92
Nakano K, Bálint E, Ashcroft M, Vousden KH. A ribonucleotide
reductase gene is a transcriptional target of p53 and p73.
Oncogene. 2000 Aug 31;19(37):4283-9
Tanaka H, Arakawa H, Yamaguchi T, Shiraishi K, Fukuda S,
Matsui K, Takei Y, Nakamura Y. A ribonucleotide reductase
gene involved in a p53-dependent cell-cycle checkpoint for
DNA damage. Nature. 2000 Mar 2;404(6773):42-9
Bergman AM, Eijk PP, Ruiz van Haperen VW, Smid K,
Veerman G, Hubeek I, van den Ijssel P, Ylstra B, Peters GJ. In
vivo induction of resistance to gemcitabine results in increased
expression of ribonucleotide reductase subunit M1 as the
major determinant. Cancer Res. 2005 Oct 15;65(20):9510-6
Guittet O, Håkansson P, Voevodskaya N, Fridd S, Gräslund A,
Arakawa H, Nakamura Y, Thelander L. Mammalian p53R2
protein forms an active ribonucleotide reductase in vitro with
the R1 protein, which is expressed both in resting cells in
response to DNA damage and in proliferating cells. J Biol
Chem. 2001 Nov 2;276(44):40647-51
van de Wiel MA, Costa JL, Smid K, Oudejans CB, Bergman
AM, Meijer GA, Peters GJ, Ylstra B. Expression microarray
analysis and oligo array comparative genomic hybridization of
acquired gemcitabine resistance in mouse colon reveals
selection for chromosomal aberrations. Cancer Res. 2005 Nov
15;65(22):10208-13
Yamaguchi T, Matsuda K, Sagiya Y, Iwadate M, Fujino MA,
Nakamura Y, Arakawa H. p53R2-dependent pathway for DNA
synthesis in a p53-regulated cell cycle checkpoint. Cancer
Res. 2001 Nov 15;61(22):8256-62
Bepler G, Kusmartseva I, Sharma S, Gautam A, Cantor A,
Sharma A, Simon G. RRM1 modulated in vitro and in vivo
efficacy of gemcitabine and platinum in non-small-cell lung
cancer. J Clin Oncol. 2006 Oct 10;24(29):4731-7
Zhao B, Bepler G. Transcript map and complete genomic
sequence for the 310 kb region of minimal allele loss on
chromosome segment 11p15.5 in non-small-cell lung cancer.
Oncogene. 2001 Dec 6;20(56):8154-64
Ceppi P, Volante M, Novello S, Rapa I, Danenberg KD, et al.
ERCC1 and RRM1 gene expressions but not EGFR are
predictive of shorter survival in advanced non-small-cell lung
cancer treated with cisplatin and gemcitabine. Ann Oncol. 2006
Dec;17(12):1818-25
Bepler G, Gautam A, McIntyre LM, Beck AF, Chervinsky DS,
Kim YC, Pitterle DM, Hyland A. Prognostic significance of
molecular genetic aberrations on chromosome segment
11p15.5 in non-small-cell lung cancer. J Clin Oncol. 2002 Mar
1;20(5):1353-60
Ferraro P, Nicolosi L, Bernardi P, Reichard P, Bianchi V.
Mitochondrial deoxynucleotide pool sizes in mouse liver and
evidence for a transport mechanism for thymidine
monophosphate. Proc Natl Acad Sci U S A. 2006 Dec
5;103(49):18586-91
Gautam A, Li ZR, Bepler G. RRM1-induced metastasis
suppression through PTEN-regulated pathways. Oncogene.
2003 Apr 10;22(14):2135-42
Gautam A, Bepler G. Suppression of lung tumor formation by
the regulatory subunit of ribonucleotide reductase. Cancer
Res. 2006 Jul 1;66(13):6497-502
Kimura T, Takeda S, Sagiya Y, Gotoh M, Nakamura Y,
Arakawa H. Impaired function of p53R2 in Rrm2b-null mice
causes severe renal failure through attenuation of dNTP pools.
Nat Genet. 2003 Aug;34(4):440-5
Håkansson P, Hofer A, Thelander L. Regulation of mammalian
ribonucleotide reduction and dNTP pools after DNA damage
and in resting cells. J Biol Chem. 2006 Mar 24;281(12):783441
Rosell R, Crino L, Danenberg K, Scagliotti G, Bepler G, Taron
M, Alberola V, Provencio M, Camps C, De Marinis F, Sanchez
JJ, Peñas R. Targeted therapy in combination with
gemcitabine in non-small cell lung cancer. Semin Oncol. 2003
Aug;30(4 Suppl 10):19-25
Kwon WS, Rha SY, Choi YH, Lee JO, Park KH, Jung JJ, Kim
TS, Jeung HC, Chung HC. Ribonucleotide reductase M1
(RRM1) 2464G>A polymorphism shows an association with
gemcitabine
chemosensitivity
in
cancer
cell
lines.
Pharmacogenet Genomics. 2006 Jun;16(6):429-38
Xue L, Zhou B, Liu X, Qiu W, Jin Z, Yen Y. Wild-type p53
regulates human ribonucleotide reductase by protein-protein
interaction with p53R2 as well as hRRM2 subunits. Cancer
Res. 2003 Mar 1;63(5):980-6
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
477
RRM1 (ribonucleotide reductase M1)
Freeman SN, Bepler G
Nordlund P, Reichard P. Ribonucleotide reductases. Annu Rev
Biochem. 2006;75:681-706
Ohhashi S, Ohuchida K, Mizumoto K, Fujita H, Egami T, Yu J,
Toma H, Sadatomi S, Nagai E, Tanaka M. Down-regulation of
deoxycytidine kinase enhances acquired resistance to
gemcitabine in pancreatic cancer. Anticancer Res. 2008 JulAug;28(4B):2205-12
Oguri T, Achiwa H, Sato S, Bessho Y, Takano Y, Miyazaki M,
Muramatsu H, Maeda H, Niimi T, Ueda R. The determinants of
sensitivity and acquired resistance to gemcitabine differ in nonsmall cell lung cancer: a role of ABCC5 in gemcitabine
sensitivity. Mol Cancer Ther. 2006 Jul;5(7):1800-6
Ohtaka K, Kohya N, Sato K, Kitajima Y, Ide T, Mitsuno M,
Miyazaki K. Ribonucleotide reductase subunit M1 is a possible
chemoresistance marker to gemcitabine in biliary tract
carcinoma. Oncol Rep. 2008 Aug;20(2):279-86
Bourdon A, Minai L, Serre V, Jais JP, Sarzi E, Aubert S, et al.
Mutation of RRM2B, encoding p53-controlled ribonucleotide
reductase (p53R2), causes severe mitochondrial DNA
depletion. Nat Genet. 2007 Jun;39(6):776-80
Pontarin G, Fijolek A, Pizzo P, Ferraro P, Rampazzo C,
Pozzan T, Thelander L, Reichard PA, Bianchi V.
Ribonucleotide reduction is a cytosolic process in mammalian
cells independently of DNA damage. Proc Natl Acad Sci U S A.
2008 Nov 18;105(46):17801-6
Nakahira S, Nakamori S, Tsujie M, Takahashi Y, Okami J, et
al. Involvement of ribonucleotide reductase M1 subunit
overexpression in gemcitabine resistance of human pancreatic
cancer. Int J Cancer. 2007 Mar 15;120(6):1355-63
Pontarin G, Fijolek A, Pizzo P, Ferraro P, Rampazzo C,
Pozzan T, Thelander L, Reichard PA, Bianchi V.
Ribonucleotide reduction is a cytosolic process in mammalian
cells independently of DNA damage. Proc Natl Acad Sci U S A.
2008 Nov 18;105(46):17801-6
Nakano Y, Tanno S, Koizumi K, Nishikawa T, Nakamura K,
Minoguchi M, Izawa T, Mizukami Y, Okumura T, Kohgo Y.
Gemcitabine chemoresistance and molecular markers
associated with gemcitabine transport and metabolism in
human pancreatic cancer cells. Br J Cancer. 2007 Feb
12;96(3):457-63
Souglakos J, Boukovinas I, Taron M, Mendez P, et al.
Ribonucleotide reductase subunits M1 and M2 mRNA
expression levels and clinical outcome of lung adenocarcinoma
patients treated with docetaxel/gemcitabine. Br J Cancer. 2008
May 20;98(10):1710-5
Tooker P, Yen WC, Ng SC, Negro-Vilar A, Hermann TW.
Bexarotene (LGD1069, Targretin), a selective retinoid X
receptor agonist, prevents and reverses gemcitabine
resistance in NSCLC cells by modulating gene amplification.
Cancer Res. 2007 May 1;67(9):4425-33
Akita H, Zheng Z, Takeda Y, Chiwan K, Kittaka N, Kobayashi
S, Marubashi S, Takemasa I, Nagano H, Dono K, Nakamori S,
Monden M, Mori M, Doki Y, Bepler G.. Significance of RRM1
and
ERCC1
expression
in
resectable
pancreatic
adenocarcinoma. Oncogene. 2009: in press.
Zheng Z, Chen T, Li X, Haura E, Sharma A, Bepler G. DNA
synthesis and repair genes RRM1 and ERCC1 in lung cancer.
N Engl J Med. 2007 Feb 22;356(8):800-8
Harshman LC, Bepler G, Zheng Z, Higgins JP, Allen GI,
Srinivas S.. RRM1 expression in muscle invasive locally
advanced urothelial cancer may correlate with age. Proc Am
Soc Clin Oncol. 2009: in press.
Zhou SF. ERCC1 and non-small-cell lung cancer. N Engl J
Med. 2007 Jun 14;356(24):2540; author reply 2540-1
Bepler G, Sommers KE, Cantor A, Li X, Sharma A, et al.
Clinical efficacy and predictive molecular markers of
neoadjuvant gemcitabine and pemetrexed in resectable nonsmall cell lung cancer. J Thorac Oncol. 2008 Oct;3(10):1112-8
Metro G, Zheng Z, Fabi A, Schell M, Antoniani B, Mottolese M,
Monteiro AN, Vici P, Lara Rivera S, Boulware D, Cognetti F,
Bepler G.. In situ protein expression of RRM1, ERCC1 and
BRCA1 in metastatic breast cancer patients treated with
gemcitabine-based chemotherapy. Cancer Invest. 2009: in
press.
Boukovinas I, Papadaki C, Mendez P, Taron M, Mavroudis D,
et al. Tumor BRCA1, RRM1 and RRM2 mRNA expression
levels and clinical response to first-line gemcitabine plus
docetaxel in non-small-cell lung cancer patients. PLoS One.
2008;3(11):e3695
Reynolds C, Obasaju C, Schell MJ, Li X, Zheng Z, et al.
Randomized
phase
III
trial
of
gemcitabine-based
chemotherapy with in situ RRM1 and ERCC1 protein levels for
response prediction in non-small-cell lung cancer. J Clin Oncol.
2009: in press.
Chang L, Zhou B, Hu S, Guo R, Liu X, Jones SN, Yen Y. ATMmediated serine 72 phosphorylation stabilizes ribonucleotide
reductase small subunit p53R2 protein against MDM2 to DNA
damage. Proc Natl Acad Sci U S A. 2008 Nov
25;105(47):18519-24
This article should be referenced as such:
Freeman SN, Bepler G. RRM1 (ribonucleotide reductase M1).
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5):473-478.
Kim SO, Jeong JY, Kim MR, Cho HJ, Ju JY, Kwon YS, Oh IJ,
Kim KS, Kim YI, Lim SC, Kim YC. Efficacy of gemcitabine in
patients with non-small cell lung cancer according to promoter
polymorphisms of the ribonucleotide reductase M1 gene. Clin
Cancer Res. 2008 May 15;14(10):3083-8
Atlas Genet Cytogenet Oncol Haematol. 2010; 14(5)
478