Download Supplementary Table S9 (doc 396K)

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
Table S9. Genes that may mediate transformation by NUP98 fusions.
Upregulated Genes
Homeobox
Several genes that encode homeodomain-containing transcription factors
genes
are upregulated by NUP98 fusions. These include the clustered (type I)
HOX genes HOXA3, HOXA5, HOXA6, HOXA7, HOXA9, HOXB3,
HOXB5, HOXB6, and HOXB7 as well as the non-clustered (type II)
homeobox genes, MEIS1 and PBX3. Homeobox genes play important
roles in normal hematopoiesis and in leukemogenesis (1, 2). Several are
expressed early in hematopoiesis and downregulated with differentiation
(3). Overexpression of HOXA9 in mouse bone marrow causes AML that
is accelerated by coexpression of MEIS1 (4). MEIS1 accelerates the
induction of AML by NUP98-HOXA9 in mice (4). Several homeobox
genes including HOXA9, HOXA7, and MEIS1 are frequently
overexpressed in human AML and are associated with worse prognosis
(5-9). Homeobox gene overexpression is believed to mediate the
leukemogenic effects of MLL fusions (10). The induction of several
homeobox genes by both NUP98-HOXA9 and NUP98-DDX10 suggests
that homeobox genes play an important role in leukemic transformation
by NUP98 fusions. This notion is supported by data showing that
homeobox genes are upregulated in murine bone marrow cells
expressing other leukemogenic NUP98 fusions such as NUP98HOXD13, NUP98-PMX1, NUP98-NSD1, and NUP98-HHEX (11-15).
EGR1
This is an immediate-early gene that encodes a zinc finger transcription
factor (16). It is upregulated in human AML with monocytic differentiation
(17) and in prostate cancer (18). It is also upregulated in a mouse model
of acute promyelocytic leukemia (19) and there is evidence that it
mediates the proliferative effects of erythropoietin on mouse
erythroblasts (20). These data suggest that upregulation of EGR1 by
NUP98 fusions may contribute to the leukemic transformation of primary
human cells, particularly to the increased numbers of erythroid cells
observed.
SOX4
This is a transcription factor that contains an HMG DNA-binding domain
(21). It can cooperate with either MEF2C or EVI1 to induce AML in mice
(22, 23) and is induced by the AML-associated RUNX1-ETO fusion
oncoprotein in primary human hematopoietic cells (24).
MYCN
This is a basic helix-loop-helix/leucine zipper (bHLH/LZ) transcription
factor that is well known for its amplification and overexpression in
neuroblastoma and other pediatric tumors (25). MYCN is also amplified
and overexpressed in lymphomas (26, 27). More recently, it was
reported that MYCN is overexpressed in many cases of pediatric AML
and that its overexpression in mice leads to the development of AML
(28). Therefore upregulation of MYCN may contribute to leukemic
transformation by NUP98 fusions.
ZNF521
Also known as EHZF, this is a zinc finger protein that is expressed in
primitive human CD34+ hematopoietic cells and declines with
differentiation (29). It is expressed in most cases of AML (29). It interacts
with of SMAD1, SMAD4, and GATA-1 and regulates their transcriptional
activity (29, 30). There is evidence that it inhibits erythroid differentiation
by inhibiting the transcriptional activity of GATA-1 (30).
HEY1
PTGS2
REN
This is a b-HLH protein also known as HERP2 that acts as a
transcriptional repressor. It is a mediator in the Notch signaling pathway
(31) and is implicated in the pathogenesis of tumors of the brain, lung,
pancreas, bone, skin, and kidney (32-37). Notch signaling is believed to
be important in the self-renewal of hematopoietic stem cells (38) and in
the pathogenesis of hematologic malignancies, particularly Tlymphoblastic leukemia (39). These data suggest that upregulation of
HEY1 may contribute to the long-term proliferation and increased
numbers of LTC-ICs induced by NUP98 fusions. HEY1 plays a role in
erythroid differentiation: in a mouse cell line, Notch signaling associated
with HEY1 upregulation increased the numbers of erythroid cells (40),
and upregulation of HEY1 by JUN in primary human hematopoietic cells
was associated with a block in erythroid differentiation (41). The
somewhat different results obtained in these two studies may reflect the
different experimental systems used, but the data nevertheless suggest
that the increased numbers of erythroid cells and the disrupted erythroid
differentiation that we observed in primary human cells expressing
NUP98 fusions may be mediated at least in part by upregulation of
HEY1.
This
gene
encodes prostaglandin-endoperoxide
synthase
2
(prostaglandin G/H synthase 2), better known as cyclooxygenase-2
(COX-2), which converts arachidonic acid to prostaglandins G2 and H2,
precursors of a number of different prostaglandins (42). COX-2 is
overexpressed in many tumors resulting in overproduction of
prostaglandins, including PGE2, which is thought to be important in
carcinogenesis (42). Many types of cancer overexpress COX-2 and its
inhibition with drugs such as aspirin can be used in the prevention and
treatment of cancer (43). A number of hematopoietic malignancies,
including chronic myleogenous leukemia, chronic lymphocytic leukemia,
lymphomas, and myeloma, show overexpression of COX-2, which is
associated with a worse prognosis (44). There is evidence that the use
of aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) is
associated with a lower incidence of lymphomas and acute leukemia
(45). In particular, NSAID use can reduce the risk of AML, particularly
the FAB M2 subtype (AML with maturation) (46), which is the most
common subtype in cases with NUP98 gene rearrangements (47). The
upregulation of PTGS2 by NUP98 fusions may contribute to the worse
prognosis of AML associated with NUP98 gene rearrangements, and
raises the possibility that COX-2 blockade could help in the treatment of
these leukemias.
This gene encodes renin, a constituent of the renin-angiotensin system
(RAS) that converts angiotensinogen to angiotensin I, which in turn is
converted by angiotensin-converting enzyme (ACE) to angiotensin II, a
regulator of blood pressure and electrolyte balance (48). In addition to
this role, the RAS may play an important role in hematopoiesis and AML
(49). Indeed, angiotensin II has been shown to increase the proliferation
of both primitive and committed hematopoietic progenitors (50, 51).
Blasts from many AML patients express renin while normal bone marrow
cells do not (52-54); in addition, angiotensinogen and ACE are also
expressed in bone marrow from AML patients (55). Thus it is possible
that the enhanced expression of renin is a part of the mechanism for
ANGPT1
RYK
STYK1
TMOD1
IDO2
uncontrolled cell growth in some leukemias. Interestingly, drugs that
inhibit the RAS system, including ACE inhibitors and losartan, have been
shown to inhibit growth and induce apoptosis in AML cell lines in vitro
(56). These data raise the possibility that such drugs may be useful in
the treatment of AML associated with NUP98 gene rearrangements.
Angiopoietin-1 is an angiogenic growth factor that is produced by
leukemic cells from AML patients (57). It is co-expressed with its
receptor, Tie2 (TEK), in AML cells and is thought to act as an autocrine
and paracrine factor that enhances angiogenesis and the growth of AML
cells (58-60).
This is a tyrosine kinase with an aberrant catalytic domain that acts as a
receptor in Wnt signaling (61). The WNT signaling pathway, particularly
the Wnt3a ligand, plays an important role in maintaining hematopoietic
stem cell function (62). RYK interacts with Wnt3a and mediates Wnt
signaling (63). It has been identified in maturing hematopoietic cells in
mice (64). Finally, RYK is overexpressed in ovarian cancer and is
capable of transforming mouse fibroblasts (65, 66).
Also known as NOK, this is a putative protein kinase that transforms cells
in vitro and promotes their tumorigenesis and metastasis in mice (67,
68). It is overexpressed in breast and lung cancer (69, 70).
This gene encodes tropomodulin 1, a member of the tropomodulin family
of proteins that cap the slow-growing pointed end of actin filaments (71).
Tropomodulin 1 is expressed in erythroid cells (71), and its upregulation
by NUP98 fusions likely reflects the increased numbers of erythroid cells
observed.
The first step in the catabolism of tryptophan is catalyzed by two related
indoleamine 2,3-dioxygenase enzymes, IDO and IDO2 (72). IDO is
expressed by many human tumors and by dendritic cells and is thought
to facilitate the escape of tumors from immune surveillance (73). An
inhibitor of indoleamine 2,3-dioxygenase enzymes, 1-methyl-tryptophan,
shows antitumor activity in mice (73). Interestingly, the D-isomer of 1methyl tryptophan, which preferentially inhibits IDO2, shows higher
antitumor activity than the L-isomer, which targets IDO (74, 75).
Downregulated genes:
RAP1A
This gene a small GTPase of the Ras family that was identified as a
suppressor of the transforming activity of oncogenic Ras in NIH/3T3 cells
(76). It was subsequently found to potentiate the functions of Ras in some
cells and to antagonize them in others (77). RAP1A functions in integrinmediated adhesion and signaling and in the MAP kinase cascade (77). It
plays various roles in the proliferation and differentiation of hematopoietic
cells (78). For example, megakaryocytic, monocytic, and lymphoid
differentiation of cell lines is associated with RAP1A induction (79, 80).
Maturation of megakaryocytes derived from human cord blood cells is also
associated with induction of RAP1A (81).
Thrombopoietin-induced
megakaryocytic differentiation is mediated by sustained activation of the
ERK/MAPK pathway mediated by RAP1A activation and inhibition of
megakaryocytic differentiation by stromal contact is associated with a
block in RAP1A activation (80, 82). Finally, RAP1A is activated by
erythropoietin (83), raising the possibility that it plays a role in erythroid
FCGR2C
CLEC7A
ALOX5
FPR3
A2M
ELA2
GADD45B
MS4A3
TRIM35
ZFHX3
differentiation. Based on these data, the repression of RAP1A expression
by both NUP98 fusions may play a role in their inhibition of both erythroid
and myeloid differentiation.
This is an isoform of FCGR2, also known as CD32. It is expressed on
myelomonocytic cells and is upregulated during myeloid differentiation
(84-87). Its downregulation by NUP98 fusions is consistent with the
observed disruption of myeloid differentiation.
This gene, also known as Dectin-1, encodes a beta-glucan receptor that is
expressed on myeloid cells including monocytes/macrophages,
neutrophils, and eosinophils (88-90). Its repression by NUP98 fusions
may reflect disruption of myeloid differentiation.
This is a member of the lipoxygenase gene family that plays a role in the
synthesis of leukotrienes from arachidonic acid. It is transcriptionally
upregulated during myeloid differentiation (91-93). Its repression by
NUP98 fusions likely reflects disrupted myeloid differentiation.
This is a member of the family of formyl peptide receptors; it is also known
as FPRL2 and expressed in monocytes (94). Its repression by NUP98
fusions may reflect disrupted myeloid differentiation.
This gene encodes the protease inhibitor alpha-2-macroglobulin, which is
induced during the maturation of monocytes to macrophages (95, 96). Its
repression by NUP98 fusions may reflect disrupted myeloid differentiation.
This gene encodes elastase, a component of neutrophil granules (97). Its
downregulation by NUP98 fusions is consistent with their disruption of
myeloid differentiation.
Also known as MyD118, this is one of a closely related family of genes
associated with terminal myeloid differentiation and involved in the
induction of cell cycle arrest and apoptosis (98). Its early downregulation
by NUP98 fusions may contribute to blocked differentiation and increased
proliferation.
This gene, also known as HTm4, belongs to a family of proteins with 4
membrane-spanning domains and is expressed predominantly in
hematopoietic cells (99). It is involved in the regulation of the cell cycle in
hematopoietic cells where its overexpression leads to cell cycle arrest at
the G0/G1 phase (100). Repression of the myeloid oncogene AML1-ETO
in cell lines and primary AML blasts results in induction of MS4A3 (101),
indicating that the proliferative effects of AML1-ETO are mediated by
downregulation of MS4A3.
Also known as Hls5 or MAIR, this gene is thought to act as a tumor
suppressor that induces apoptosis and is associated with erythroid-tomyeloid lineage switch (102-104). Its repression by NUP98 fusions may
explain the skewing towards erythroid differentiation (Figure 3); it also
raises the possibility that the increased numbers of cells in long-term
culture (Figure 2) may be due in part to decreased apoptosis.
This gene is also known as ATBF1 and encodes two isoforms of a
transcription factor that contains multiple homeodomains and zinc fingers
(105, 106).
It is thought to act as a tumor suppressor that is
downregulated in cancers of the prostate, breast, liver, and stomach (107110). In neuronal cells, it induces differentiation and cell cycle arrest
(111). Based on these findings, it is possible that downregulation of
ZFHX3 by NUP98 fusions plays a role in their ability to block differentiation
NDRG2
RAD18
EED
GPX3
and induce proliferation in hematopoietic cells.
This is a member of the N-myc downregulated gene family. It is thought to
act as a tumor suppressor that is downregulated in many tumors including
carcinomas of the colon and thyroid, glioblastoma, and others (112-116).
Its repression by NUP98 fusions suggests a similar role as a tumor
suppressor in AML.
This gene encodes a ubiquitin ligase involved in DNA repair (117, 118).
Its downregulation by NUP98 fusions may predispose cells to further DNA
damage. For example in patients treated with topoisomerase II inhibitors,
who are susceptible to NUP98 gene rearrangements (47), such a
mechanism may lead to additional mutations that could cooperate with the
NUP98 fusion in producing AML.
This is one of the Polycomb Group (PcG) gene that act as transcriptional
repressors (119). Two Polycomb repressive complexes (PRC) have been
described: PRC1 and PRC2. EED is a component of the PRC2 complex
that methylates H3K27 and represses the expression of several HOX
genes (119, 120). Interestingly, several HOX genes including HOXA7,
HOXA9, MEIS1, and others are upregulated by NUP98 fusions
(Supplementary Tables S6 – S8). This upregulation of HOX genes may
be explained at least in part by repression of EED.
This gene encodes glutathione peroxidase 3, a selenium dependent
enzyme that plays a role in detoxifying reactive oxygen species and is
downregulated in a number of cancers including those of the prostate,
esophagus, and stomach (121-125). Its reression by NUP98 fusions
suggests that it may play a role in AML as well.
References
1.
Argiropoulos B, Humphries RK. Hox genes in hematopoiesis
leukemogenesis. Oncogene 2007 Oct 15; 26(47): 6766-6776.
and
2.
Sitwala KV, Dandekar MN, Hess JL. HOX Proteins and Leukemia. Int J Clin Exp
Pathol 2008; 1(6): 461-474.
3.
Pineault N, Helgason CD, Lawrence HJ, Humphries RK. Differential expression
of Hox, Meis1, and Pbx1 genes in primitive cells throughout murine
hematopoietic ontogeny. Exp Hematol 2002 Jan; 30(1): 49-57.
4.
Kroon E, Thorsteinsdottir U, Mayotte N, Nakamura T, Sauvageau G. NUP98HOXA9 expression in hemopoietic stem cells induces chronic and acute myeloid
leukemias in mice. Embo J 2001; 20(3): 350-361.
5.
Kawagoe H, Humphries RK, Blair A, Sutherland HJ, Hogge DE. Expression of
HOX genes, HOX cofactors, and MLL in phenotypically and functionally defined
subpopulations of leukemic and normal human hematopoietic cells. Leukemia
1999 May; 13(5): 687-698.
6.
Lawrence HJ, Rozenfeld S, Cruz C, Matsukuma K, Kwong A, Komuves L, et al.
Frequent co-expression of the HOXA9 and MEIS1 homeobox genes in human
myeloid leukemias. Leukemia 1999; 13(12): 1993-1999.
7.
Afonja O, Smith JE, Jr., Cheng DM, Goldenberg AS, Amorosi E, Shimamoto T, et
al. MEIS1 and HOXA7 genes in human acute myeloid leukemia. Leuk Res 2000;
24(10): 849-855.
8.
Drabkin HA, Parsy C, Ferguson K, Guilhot F, Lacotte L, Roy L, et al. Quantitative
HOX expression in chromosomally defined subsets of acute myelogenous
leukemia. Leukemia 2002; 16(2): 186-195.
9.
Bullinger L, Dohner K, Bair E, Frohling S, Schlenk RF, Tibshirani R, et al. Use of
gene-expression profiling to identify prognostic subclasses in adult acute myeloid
leukemia. N Engl J Med 2004 Apr 15; 350(16): 1605-1616.
10.
Dou Y, Hess JL. Mechanisms of transcriptional regulation by MLL and its
disruption in acute leukemia. Int J Hematol 2008 Jan; 87(1): 10-18.
11.
Jankovic D, Gorello P, Liu T, Ehret S, La Starza R, Desjobert C, et al.
Leukemogenic mechanisms and targets of a NUP98/HHEX fusion in acute
myeloid leukemia. Blood 2008 Jun 15; 111(12): 5672-5682.
12.
Palmqvist L, Pineault N, Wasslavik C, Humphries RK. Candidate genes for
expansion and transformation of hematopoietic stem cells by NUP98-HOX fusion
genes. PLoS ONE 2007; 2(1): e768.
13.
Pineault N, Buske C, Feuring-Buske M, Abramovich C, Rosten P, Hogge DE, et
al. Induction of acute myeloid leukemia in mice by the human leukemia-specific
fusion gene NUP98-HOXD13 in concert with Meis1. Blood 2003 Jun 1; 101(11):
4529-4538.
14.
Hirose K, Abramovich C, Argiropoulos B, Humphries RK. Leukemogenic
properties of NUP98-PMX1 are linked to NUP98 and homeodomain sequence
functions but not to binding properties of PMX1 to serum response factor.
Oncogene 2008 Oct 9; 27(46): 6056-6067.
15.
Wang GG, Cai L, Pasillas MP, Kamps MP. NUP98-NSD1 links H3K36
methylation to Hox-A gene activation and leukaemogenesis. Nat Cell Biol 2007
Jul; 9(7): 804-812.
16.
Sukhatme VP. Early transcriptional events in cell growth: the Egr family. J Am
Soc Nephrol 1990 Dec; 1(6): 859-866.
17.
Tokura Y, Shikami M, Miwa H, Watarai M, Sugamura K, Wakabayashi M, et al.
Augmented expression of P-gp/multi-drug resistance gene by all-trans retinoic
acid in monocytic leukemic cells. Leuk Res 2002 Jan; 26(1): 29-36.
18.
Abdulkadir SA. Mechanisms of prostate tumorigenesis: roles for transcription
factors Nkx3.1 and Egr1. Ann N Y Acad Sci 2005 Nov; 1059: 33-40.
19.
Yuan W, Payton JE, Holt MS, Link DC, Watson MA, DiPersio JF, et al.
Commonly dysregulated genes in murine APL cells. Blood 2007 Feb 1; 109(3):
961-970.
20.
Fang J, Menon M, Kapelle W, Bogacheva O, Bogachev O, Houde E, et al. EPO
modulation of cell-cycle regulatory genes, and cell division, in primary bone
marrow erythroblasts. Blood 2007 Oct 1; 110(7): 2361-2370.
21.
Schilham MW, Clevers H. HMG box containing transcription factors in
lymphocyte differentiation. Semin Immunol 1998 Apr; 10(2): 127-132.
22.
Du Y, Spence SE, Jenkins NA, Copeland NG. Cooperating cancer-gene
identification through oncogenic-retrovirus-induced insertional mutagenesis.
Blood 2005 Oct 1; 106(7): 2498-2505.
23.
Boyd KE, Xiao YY, Fan K, Poholek A, Copeland NG, Jenkins NA, et al. Sox4
cooperates with Evi1 in AKXD-23 myeloid tumors via transactivation of proviral
LTR. Blood 2005 Oct 4.
24.
Tonks A, Pearn L, Musson M, Gilkes A, Mills KI, Burnett AK, et al. Transcriptional
dysregulation mediated by RUNX1-RUNX1T1 in normal human progenitor cells
and in acute myeloid leukaemia. Leukemia 2007 Dec; 21(12): 2495-2505.
25.
Pession A, Tonelli R. The MYCN oncogene as a specific and selective drug
target for peripheral and central nervous system tumors. Curr Cancer Drug
Targets 2005 Jun; 5(4): 273-283.
26.
Mao X, Onadim Z, Price EA, Child F, Lillington DM, Russell-Jones R, et al.
Genomic alterations in blastic natural killer/extranodal natural killer-like T cell
lymphoma with cutaneous involvement. J Invest Dermatol 2003 Sep; 121(3):
618-627.
27.
Schwaenen C, Nessling M, Wessendorf S, Salvi T, Wrobel G, Radlwimmer B, et
al. Automated array-based genomic profiling in chronic lymphocytic leukemia:
development of a clinical tool and discovery of recurrent genomic alterations.
Proc Natl Acad Sci U S A 2004 Jan 27; 101(4): 1039-1044.
28.
Kawagoe H, Kandilci A, Kranenburg TA, Grosveld GC. Overexpression of N-Myc
rapidly causes acute myeloid leukemia in mice. Cancer Res 2007 Nov 15;
67(22): 10677-10685.
29.
Bond HM, Mesuraca M, Amodio N, Mega T, Agosti V, Fanello D, et al. Early
hematopoietic zinc finger protein-zinc finger protein 521: a candidate regulator of
diverse immature cells. Int J Biochem Cell Biol 2008; 40(5): 848-854.
30.
Matsubara E, Sakai I, Yamanouchi J, Fujiwara H, Yakushijin Y, Hato T, et al. The
role of zinc finger protein 521/early hematopoietic zinc finger protein in erythroid
cell differentiation. J Biol Chem 2009 Feb 6; 284(6): 3480-3487.
31.
Iso T, Kedes L, Hamamori Y. HES and HERP families: multiple effectors of the
Notch signaling pathway. J Cell Physiol 2003 Mar; 194(3): 237-255.
32.
Cavard C, Audebourg A, Letourneur F, Audard V, Beuvon F, Cagnard N, et al.
Gene expression profiling provides insights into the pathways involved in solid
pseudopapillary neoplasm of the pancreas. J Pathol 2009 Jan 20.
33.
Engin F, Bertin T, Ma O, Jiang MM, Wang L, Sutton RE, et al. Notch Signaling
Contributes to the Pathogenesis of Human Osteosarcomas. Hum Mol Genet
2009 Feb 19.
34.
Hulleman E, Quarto M, Vernell R, Masserdotti G, Colli E, Kros JM, et al. A role
for the transcription factor HEY1 in glioblastoma. J Cell Mol Med 2009 Jan; 13(1):
136-146.
35.
Kang S, Yang C, Luo R. Induction of CCL2 by siMAML1 through upregulation of
TweakR in melanoma cells. Biochem Biophys Res Commun 2008 Aug 8; 372(4):
629-633.
36.
Konishi J, Kawaguchi KS, Vo H, Haruki N, Gonzalez A, Carbone DP, et al.
Gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation
in human lung cancers. Cancer Res 2007 Sep 1; 67(17): 8051-8057.
37.
Lucas B, Grigo K, Erdmann S, Lausen J, Klein-Hitpass L, Ryffel GU. HNF4alpha
reduces proliferation of kidney cells and affects genes deregulated in renal cell
carcinoma. Oncogene 2005 Sep 22; 24(42): 6418-6431.
38.
Moore MA. Converging pathways in leukemogenesis and stem cell self-renewal.
Exp Hematol 2005 Jul; 33(7): 719-737.
39.
Nefedova Y, Gabrilovich D. Mechanisms and clinical prospects of Notch
inhibitors in the therapy of hematological malignancies. Drug Resist Updat 2008
Dec; 11(6): 210-218.
40.
Henning K, Schroeder T, Schwanbeck R, Rieber N, Bresnick EH, Just U.
mNotch1 signaling and erythropoietin cooperate in erythroid differentiation of
multipotent progenitor cells and upregulate beta-globin. Exp Hematol 2007 Sep;
35(9): 1321-1332.
41.
Elagib KE, Xiao M, Hussaini IM, Delehanty LL, Palmer LA, Racke FK, et al. Jun
blockade of erythropoiesis: role for repression of GATA-1 by HERP2. Mol Cell
Biol 2004 Sep; 24(17): 7779-7794.
42.
Ulrich CM, Bigler J, Potter JD. Non-steroidal anti-inflammatory drugs for cancer
prevention: promise, perils and pharmacogenetics. Nat Rev Cancer 2006 Feb;
6(2): 130-140.
43.
Harris RE. Cyclooxygenase-2 (cox-2) and the inflammogenesis of cancer.
Subcell Biochem 2007; 42: 93-126.
44.
Bernard MP, Bancos S, Sime PJ, Phipps RP. Targeting cyclooxygenase-2 in
hematological malignancies: rationale and promise. Curr Pharm Des 2008;
14(21): 2051-2060.
45.
Robak P, Smolewski P, Robak T. The role of non-steroidal anti-inflammatory
drugs in the risk of development and treatment of hematologic malignancies.
Leuk Lymphoma 2008 Aug; 49(8): 1452-1462.
46.
Pogoda JM, Katz J, McKean-Cowdin R, Nichols PW, Ross RK, Preston-Martin S.
Prescription drug use and risk of acute myeloid leukemia by French-AmericanBritish subtype: results from a Los Angeles County case-control study. Int J
Cancer 2005 Apr 20; 114(4): 634-638.
47.
Romana SP, Radford-Weiss I, Ben Abdelali R, Schluth C, Petit A, Dastugue N, et
al. NUP98 rearrangements in hematopoietic malignancies: a study of the Groupe
Francophone de Cytogenetique Hematologique. Leukemia 2006 Apr; 20(4): 696706.
48.
Dzau VJ. Circulating versus local renin-angiotensin system in cardiovascular
homeostasis. Circulation 1988 Jun; 77(6 Pt 2): I4-13.
49.
Haznedaroglu IC, Ozturk MA. Towards the understanding of the local
hematopoietic bone marrow renin-angiotensin system. Int J Biochem Cell Biol
2003 Jun; 35(6): 867-880.
50.
Mrug M, Stopka T, Julian BA, Prchal JF, Prchal JT. Angiotensin II stimulates
proliferation of normal early erythroid progenitors. J Clin Invest 1997 Nov 1;
100(9): 2310-2314.
51.
Rodgers KE, Xiong S, Steer R, diZerega GS. Effect of angiotensin II on
hematopoietic progenitor cell proliferation. Stem Cells 2000; 18(4): 287-294.
52.
Wulf GG, Jahns-Streubel G, Nobiling R, Strutz F, Hemmerlein B, Hiddemann W,
et al. Renin in acute myeloid leukaemia blasts. Br J Haematol 1998; 100(2): 335337.
53.
Inigo Sde L, Casares MT, Jorge CE, Leiza SM, Santana GS, Bravo de Laguna
SJ, et al. Relevance of renin expression by real-time PCR in acute myeloid
leukemia. Leuk Lymphoma 2006 Mar; 47(3): 409-416.
54.
Teresa Gomez Casares M, de la Iglesia S, Perera M, Lemes A, Campo C,
Gonzalez San Miguel JD, et al. Renin expression in hematological malignancies
and its role in the regulation of hematopoiesis. Leuk Lymphoma 2002 Dec;
43(12): 2377-2381.
55.
Beyazit Y, Aksu S, Haznedaroglu IC, Kekilli M, Misirlioglu M, Tuncer S, et al.
Overexpression of the local bone marrow renin-angiotensin system in acute
myeloid leukemia. J Natl Med Assoc 2007 Jan; 99(1): 57-63.
56.
De la Iglesia Inigo S, Lopez-Jorge CE, Gomez-Casares MT, Lemes Castellano
A, Martin Cabrera P, Lopez Brito J, et al. Induction of apoptosis in leukemic cell
lines treated with captopril, trandolapril and losartan: A new role in the treatment
of leukaemia for these agents. Leuk Res 2008 Nov 14.
57.
Hatfield KJ, Hovland R, Oyan AM, Kalland KH, Ryningen A, Gjertsen BT, et al.
Release of angiopoietin-1 by primary human acute myelogenous leukemia cells
is associated with mutations of nucleophosmin, increased by bone marrow
stromal cells and possibly antagonized by high systemic angiopoietin-2 levels.
Leukemia 2008 Feb; 22(2): 287-293.
58.
Hatfield K, Oyan AM, Ersvaer E, Kalland KH, Lassalle P, Gjertsen BT, et al.
Primary human acute myeloid leukaemia cells increase the proliferation of
microvascular endothelial cells through the release of soluble mediators. Br J
Haematol 2009 Jan; 144(1): 53-68.
59.
Albitar M. Angiogenesis in acute myeloid leukemia and myelodysplastic
syndrome. Acta Haematol 2001; 106(4): 170-176.
60.
Muller A, Lange K, Gaiser T, Hofmann M, Bartels H, Feller AC, et al. Expression
of angiopoietin-1 and its receptor TEK in hematopoietic cells from patients with
myeloid leukemia. Leuk Res 2002 Feb; 26(2): 163-168.
61.
Hendrickx M, Leyns L. Non-conventional Frizzled ligands and Wnt receptors. Dev
Growth Differ 2008 May; 50(4): 229-243.
62.
Malhotra S, Kincade PW. Wnt-related molecules and signaling pathway
equilibrium in hematopoiesis. Cell Stem Cell 2009 Jan 9; 4(1): 27-36.
63.
Lu W, Yamamoto V, Ortega B, Baltimore D. Mammalian Ryk is a Wnt coreceptor
required for stimulation of neurite outgrowth. Cell 2004 Oct 1; 119(1): 97-108.
64.
Simoneaux DK, Fletcher FA, Jurecic R, Shilling HG, Van NT, Patel P, et al. The
receptor tyrosine kinase-related gene (ryk) demonstrates lineage and stagespecific expression in hematopoietic cells. J Immunol 1995 Feb 1; 154(3): 11571166.
65.
Katso RM, Manek S, Biddolph S, Whittaker R, Charnock MF, Wells M, et al.
Overexpression of H-Ryk in mouse fibroblasts confers transforming ability in vitro
and in vivo: correlation with up-regulation in epithelial ovarian cancer. Cancer
Res 1999 May 15; 59(10): 2265-2270.
66.
Wang XC, Katso R, Butler R, Hanby AM, Poulsom R, Jones T, et al. H-RYK, an
unusual receptor kinase: isolation and analysis of expression in ovarian cancer.
Mol Med 1996 Mar; 2(2): 189-203.
67.
Liu L, Yu XZ, Li TS, Song LX, Chen PL, Suo TL, et al. A novel protein tyrosine
kinase NOK that shares homology with platelet- derived growth factor/fibroblast
growth factor receptors induces tumorigenesis and metastasis in nude mice.
Cancer Res 2004 May 15; 64(10): 3491-3499.
68.
Ye X, Ji C, Huang Q, Cheng C, Tang R, Xu J, et al. Isolation and characterization
of a human putative receptor protein kinase cDNA STYK1. Mol Biol Rep 2003
Jun; 30(2): 91-96.
69.
Amachika T, Kobayashi D, Moriai R, Tsuji N, Watanabe N. Diagnostic relevance
of overexpressed mRNA of novel oncogene with kinase-domain (NOK) in lung
cancers. Lung Cancer 2007 Jun; 56(3): 337-340.
70.
Moriai R, Kobayashi D, Amachika T, Tsuji N, Watanabe N. Diagnostic relevance
of overexpressed NOK mRNA in breast cancer. Anticancer Res 2006 Nov-Dec;
26(6C): 4969-4973.
71.
Fischer RS, Fowler VM. Tropomodulins: life at the slow end. Trends Cell Biol
2003 Nov; 13(11): 593-601.
72.
Ball HJ, Yuasa HJ, Austin CJ, Weiser S, Hunt NH. Indoleamine 2,3-dioxygenase2; a new enzyme in the kynurenine pathway. Int J Biochem Cell Biol 2009 Mar;
41(3): 467-471.
73.
Prendergast GC. Immune escape as a fundamental trait of cancer: focus on IDO.
Oncogene 2008 Jun 26; 27(28): 3889-3900.
74.
Hou DY, Muller AJ, Sharma MD, DuHadaway J, Banerjee T, Johnson M, et al.
Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1methyl-tryptophan correlates with antitumor responses. Cancer Res 2007 Jan 15;
67(2): 792-801.
75.
Metz R, Duhadaway JB, Kamasani U, Laury-Kleintop L, Muller AJ, Prendergast
GC. Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target
of the antitumor indoleamine 2,3-dioxygenase inhibitory compound D-1-methyltryptophan. Cancer Res 2007 Aug 1; 67(15): 7082-7087.
76.
Kitayama H, Sugimoto Y, Matsuzaki T, Ikawa Y, Noda M. A ras-related gene with
transformation suppressor activity. Cell 1989 Jan 13; 56(1): 77-84.
77.
Stork PJ. Does Rap1 deserve a bad Rap? Trends Biochem Sci 2003 May; 28(5):
267-275.
78.
Stork PJ, Dillon TJ. Multiple roles of Rap1 in hematopoietic cells: complementary
versus antagonistic functions. Blood 2005 Nov 1; 106(9): 2952-2961.
79.
Adachi M, Ryo R, Yoshida A, Sugano W, Yasunaga M, Saigo K, et al. Induction
of smg p21/rap1A p21/krev-1 p21 gene expression during phorbol ester-induced
differentiation of a human megakaryocytic leukemia cell line. Oncogene 1992
Feb; 7(2): 323-329.
80.
Garcia J, de Gunzburg J, Eychene A, Gisselbrecht S, Porteu F. Thrombopoietinmediated sustained activation of extracellular signal-regulated kinase in UT7-Mpl
cells requires both Ras-Raf-1- and Rap1-B-Raf-dependent pathways. Mol Cell
Biol 2001 Apr; 21(8): 2659-2670.
81.
Balduini A, Pecci A, Lova P, Arezzi N, Marseglia C, Bellora F, et al. Expression,
activation, and subcellular localization of the Rap1 GTPase in cord blood-derived
human megakaryocytes. Exp Cell Res 2004 Oct 15; 300(1): 84-93.
82.
Delehanty LL, Mogass M, Gonias SL, Racke FK, Johnstone B, Goldfarb AN.
Stromal inhibition of megakaryocytic differentiation is associated with blockade of
sustained Rap1 activation. Blood 2003 Mar 1; 101(5): 1744-1751.
83.
Arai A, Nosaka Y, Kanda E, Yamamoto K, Miyasaka N, Miura O. Rap1 is
activated by erythropoietin or interleukin-3 and is involved in regulation of beta1
integrin-mediated hematopoietic cell adhesion. J Biol Chem 2001 Mar 30;
276(13): 10453-10462.
84.
Baer MR, Stewart CC, Lawrence D, Arthur DC, Mrozek K, Strout MP, et al. Acute
myeloid leukemia with 11q23 translocations: myelomonocytic immunophenotype
by multiparameter flow cytometry. Leukemia 1998 Mar; 12(3): 317-325.
85.
Barber N, Belov L, Christopherson RI. All-trans retinoic acid induces different
immunophenotypic changes on human HL60 and NB4 myeloid leukaemias. Leuk
Res 2008 Feb; 32(2): 315-322.
86.
Bassoe CF, Halstensen A, Bruserud O. Functional differentiation of acute
myeloid leukaemia blast cells. APMIS 1999 Nov; 107(11): 1023-1033.
87.
Wightman J, Roberson MS, Lamkin TJ, Varvayanis S, Yen A. Retinoic acidinduced growth arrest and differentiation: retinoic acid up-regulates CD32 (Fc
gammaRII) expression, the ectopic expression of which retards the cell cycle.
Mol Cancer Ther 2002 May; 1(7): 493-506.
88.
Ferwerda G, Meyer-Wentrup F, Kullberg BJ, Netea MG, Adema GJ. Dectin-1
synergizes with TLR2 and TLR4 for cytokine production in human primary
monocytes and macrophages. Cell Microbiol 2008 Oct; 10(10): 2058-2066.
89.
Taylor PR, Brown GD, Reid DM, Willment JA, Martinez-Pomares L, Gordon S, et
al. The beta-glucan receptor, dectin-1, is predominantly expressed on the surface
of cells of the monocyte/macrophage and neutrophil lineages. J Immunol 2002
Oct 1; 169(7): 3876-3882.
90.
Willment JA, Marshall AS, Reid DM, Williams DL, Wong SY, Gordon S, et al. The
human beta-glucan receptor is widely expressed and functionally equivalent to
murine Dectin-1 on primary cells. Eur J Immunol 2005 May; 35(5): 1539-1547.
91.
Seuter S, Vaisanen S, Radmark O, Carlberg C, Steinhilber D. Functional
characterization of vitamin D responding regions in the human 5-Lipoxygenase
gene. Biochim Biophys Acta 2007 Jul; 1771(7): 864-872.
92.
Steinhilber D, Radmark O, Samuelsson B. Transforming growth factor beta
upregulates 5-lipoxygenase activity during myeloid cell maturation. Proc Natl
Acad Sci U S A 1993 Jul 1; 90(13): 5984-5988.
93.
Uhl J, Klan N, Rose M, Entian KD, Werz O, Steinhilber D. The 5-lipoxygenase
promoter is regulated by DNA methylation. J Biol Chem 2002 Feb 8; 277(6):
4374-4379.
94.
Migeotte I, Communi D, Parmentier M. Formyl peptide receptors: a promiscuous
subfamily of G protein-coupled receptors controlling immune responses.
Cytokine Growth Factor Rev 2006 Dec; 17(6): 501-519.
95.
Ganter U, Bauer J, Majello B, Gerok W, Ciliberto G. Characterization of
mononuclear-phagocyte terminal maturation by mRNA phenotyping using a set
of cloned cDNA probes. Eur J Biochem 1989 Nov 6; 185(2): 291-296.
96.
Saksela O, Hovi T, Vaheri A. Urokinase-type plasminogen activator and its
inhibitor secreted by cultured human monocyte-macrophages. J Cell Physiol
1985 Jan; 122(1): 125-132.
97.
Borregaard N, Cowland JB. Granules of the human
polymorphonuclear leukocyte. Blood 1997; 89(10): 3503-3521.
98.
Liebermann DA, Hoffman B. Myeloid differentiation (MyD) primary response
genes in hematopoiesis. Blood Cells Mol Dis 2003 Sep-Oct; 31(2): 213-228.
99.
Adra CN, Lelias JM, Kobayashi H, Kaghad M, Morrison P, Rowley JD, et al.
Cloning of the cDNA for a hematopoietic cell-specific protein related to CD20 and
the beta subunit of the high-affinity IgE receptor: evidence for a family of proteins
with four membrane-spanning regions. Proc Natl Acad Sci U S A 1994 Oct 11;
91(21): 10178-10182.
100.
Donato JL, Ko J, Kutok JL, Cheng T, Shirakawa T, Mao XQ, et al. Human HTm4
is a hematopoietic cell cycle regulator. J Clin Invest 2002 Jan; 109(1): 51-58.
101.
Dunne J, Cullmann C, Ritter M, Soria NM, Drescher B, Debernardi S, et al.
siRNA-mediated AML1/MTG8 depletion affects differentiation and proliferationassociated gene expression in t(8;21)-positive cell lines and primary AML blasts.
Oncogene 2006 Oct 5; 25(45): 6067-6078.
102.
Endersby R, Majewski IJ, Winteringham L, Beaumont JG, Samuels A, Scaife R,
et al. Hls5 regulated erythroid differentiation by modulating GATA-1 activity.
Blood 2008 Feb 15; 111(4): 1946-1950.
103.
Kimura F, Suzu S, Nakamura Y, Nakata Y, Yamada M, Kuwada N, et al. Cloning
and characterization of a novel RING-B-box-coiled-coil protein with apoptotic
function. J Biol Chem 2003 Jul 4; 278(27): 25046-25054.
104.
Lalonde JP, Lim R, Ingley E, Tilbrook PA, Thompson MJ, McCulloch R, et al.
HLS5, a novel RBCC (ring finger, B box, coiled-coil) family member isolated from
a hemopoietic lineage switch, is a candidate tumor suppressor. J Biol Chem
2004 Feb 27; 279(9): 8181-8189.
105.
Miura Y, Tam T, Ido A, Morinaga T, Miki T, Hashimoto T, et al. Cloning and
characterization of an ATBF1 isoform that expresses in a neuronal differentiationdependent manner. J Biol Chem 1995 Nov 10; 270(45): 26840-26848.
neutrophilic
106.
Morinaga T, Yasuda H, Hashimoto T, Higashio K, Tamaoki T. A human alphafetoprotein enhancer-binding protein, ATBF1, contains four homeodomains and
seventeen zinc fingers. Mol Cell Biol 1991 Dec; 11(12): 6041-6049.
107.
Cho YG, Song JH, Kim CJ, Lee YS, Kim SY, Nam SW, et al. Genetic alterations
of the ATBF1 gene in gastric cancer. Clin Cancer Res 2007 Aug 1; 13(15 Pt 1):
4355-4359.
108.
Kai K, Zhang Z, Yamashita H, Yamamoto Y, Miura Y, Iwase H. Loss of
heterozygosity at the ATBF1-A locus located in the 16q22 minimal region in
breast cancer. BMC Cancer 2008; 8: 262.
109.
Kim CJ, Song JH, Cho YG, Cao Z, Lee YS, Nam SW, et al. Down-regulation of
ATBF1 is a major inactivating mechanism in hepatocellular carcinoma.
Histopathology 2008 Apr; 52(5): 552-559.
110.
Lange EM, Beebe-Dimmer JL, Ray AM, Zuhlke KA, Ellis J, Wang Y, et al.
Genome-wide linkage scan for prostate cancer susceptibility from the University
of Michigan Prostate Cancer Genetics Project: suggestive evidence for linkage at
16q23. Prostate 2009 Mar 1; 69(4): 385-391.
111.
Jung CG, Kim HJ, Kawaguchi M, Khanna KK, Hida H, Asai K, et al. Homeotic
factor ATBF1 induces the cell cycle arrest associated with neuronal
differentiation. Development 2005 Dec; 132(23): 5137-5145.
112.
Kim YJ, Yoon SY, Kim JT, Choi SC, Lim JS, Kim JH, et al. NDRG2 suppresses
cell proliferation through down-regulation of AP-1 activity in human colon
carcinoma cells. Int J Cancer 2009 Jan 1; 124(1): 7-15.
113.
Liu N, Wang L, Li X, Yang Q, Liu X, Zhang J, et al. N-Myc downstream-regulated
gene 2 is involved in p53-mediated apoptosis. Nucleic Acids Res 2008 Sep;
36(16): 5335-5349.
114.
Tepel M, Roerig P, Wolter M, Gutmann DH, Perry A, Reifenberger G, et al.
Frequent promoter hypermethylation and transcriptional downregulation of the
NDRG2 gene at 14q11.2 in primary glioblastoma. Int J Cancer 2008 Nov 1;
123(9): 2080-2086.
115.
Yao L, Zhang J, Liu X. NDRG2: a Myc-repressed gene involved in cancer and
cell stress. Acta Biochim Biophys Sin (Shanghai) 2008 Jul; 40(7): 625-635.
116.
Zhao H, Zhang J, Lu J, He X, Chen C, Li X, et al. Reduced expression of N-Myc
downstream-regulated gene 2 in human thyroid cancer. BMC Cancer 2008; 8:
303.
117.
Saberi A, Hochegger H, Szuts D, Lan L, Yasui A, Sale JE, et al. RAD18 and
poly(ADP-ribose) polymerase independently suppress the access of
nonhomologous end joining to double-strand breaks and facilitate homologous
recombination-mediated repair. Mol Cell Biol 2007 Apr; 27(7): 2562-2571.
118.
Shiomi N, Mori M, Tsuji H, Imai T, Inoue H, Tateishi S, et al. Human RAD18 is
involved in S phase-specific single-strand break repair without PCNA
monoubiquitination. Nucleic Acids Res 2007; 35(2): e9.
119.
Valk-Lingbeek ME, Bruggeman SW, van Lohuizen M. Stem cells and cancer; the
polycomb connection. Cell 2004 Aug 20; 118(4): 409-418.
120.
Cao R, Zhang Y. SUZ12 is required for both the histone methyltransferase
activity and the silencing function of the EED-EZH2 complex. Mol Cell 2004 Jul 2;
15(1): 57-67.
121.
Jee CD, Kim MA, Jung EJ, Kim J, Kim WH. Identification of genes epigenetically
silenced by CpG methylation in human gastric carcinoma. Eur J Cancer 2009
Feb 3.
122.
Lee OJ, Schneider-Stock R, McChesney PA, Kuester D, Roessner A, Vieth M, et
al. Hypermethylation and loss of expression of glutathione peroxidase-3 in
Barrett's tumorigenesis. Neoplasia 2005 Sep; 7(9): 854-861.
123.
Lodygin D, Epanchintsev A, Menssen A, Diebold J, Hermeking H. Functional
epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res
2005 May 15; 65(10): 4218-4227.
124.
Peng DF, Razvi M, Chen H, Washington K, Roessner A, Schneider-Stock R, et
al. DNA hypermethylation regulates the expression of members of the Mu-class
glutathione S-transferases and glutathione peroxidases in Barrett's
adenocarcinoma. Gut 2009 Jan; 58(1): 5-15.
125.
Yu YP, Yu G, Tseng G, Cieply K, Nelson J, Defrances M, et al. Glutathione
peroxidase 3, deleted or methylated in prostate cancer, suppresses prostate
cancer growth and metastasis. Cancer Res 2007 Sep 1; 67(17): 8043-8050.