Download Gene Section RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)

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
RAF1 (v-raf-1 murine leukemia viral oncogene
homolog 1)
Max Cayo, David Yu Greenblatt, Muthusamy Kunnimalaiyaan, Herbert Chen
Endocrine Cancer Disease Group, University of Wisconsin Paul P. Carbone Comprehensive Cancer Center,
H4/750 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792, USA
Published in Atlas Database: March 2007
Online updated version: http://AtlasGeneticsOncology.org/Genes/RAF1ID42032ch3p25.html
DOI: 10.4267/2042/38452
This work is licensed under a Creative Commons Attribution-Non-commercial-No Derivative Works 2.0 France Licence.
© 2007 Atlas of Genetics and Cytogenetics in Oncology and Haematology
encoding for the serine/threonine kinase domain) in the
C terminus. The RAF proteins exhibit complex
regulation involving numerous phosphorylation sites
throughout the proteins. Despite constitutional
similarity, the Raf isoforms have been shown to carry
out non-redundant functions, implying that they are
distinct.
RAF-1 (C-RAF-1): 72-74 kDa.
Note: A-RAF: about 68 kDa.
Note: B-RAF (which undergoes alternate splicing):
ranges from 75 to 100 kDa.
Identity
Hugo: RAF1
Other names: CRAF; Raf-1; c-Raf
Location: 3p25
DNA/RNA
Note: History and Nomenclature:
c-Raf-1 was the first successfully cloned functional
human homolog of the v-Raf gene, and thus the gene
product of c-Raf-1 has historically been referred to in
the literature simply as Raf-1. Subsequently, B-Raf and
A-Raf-1 paralogues (BRAF, located in Xq13 and
ARAF, located in Xp11) were discovered. A suitable
nomenclature is as follows: A-RAF, B-RAF, and CRAF for the functional human proteins and A-RAF, BRAF, and C-RAF for the corresponding genes; a-raf, braf, and c-raf for the murine proteins and A-Raf, B-Raf,
and C-Raf for the corresponding genes. Raf-1 (or RAF1) is generally taken to mean C-RAF-1 but could apply
to A-RAF-1 equally. Here, RAF-1 will be taken to
mean C-RAF-1 (RAF-1 = C-RAF-1, etc.).
Expression
C-RAF (RAF-1) and A-RAF mRNA is expressed
ubiquitously. A-RAF mRNA is highly expressed in
urogenital organs. B-RAF is expressed in a wide range
of tissues, but most substantially in neuronal tissues.
Localisation
Cytosolic.
Function
RAF proteins are part of the conserved MAPK
(mitogen-activated protein kinase)/ERK (extracellular
signal-regulated kinase) signaling cascade between the
cell surface and the nucleus. RAF is regulated by the
upstream RAS family of small G proteins. RAS is
predominantly located on the inner leaflet of the plasma
membrane and is functionally activated by GTPbinding. Binding of various extracellular ligands such
as growth factors and hormones activates RAS and
subsequently RAF proteins. RAS binds directly to the
N-terminal regulatory domain or RAF (the RAS
binding domain (RBD)). RAS interacts secondarily
with the cysteine-rich domain (CRD) on CR1 of RAF.
RAS-RAF binding can be affected by 14-3-3 proteins
and other scaffold/adaptor proteins kinase suppressor of
Description
C-RAF (RAF-1, C-RAF-1) encompasses 80,570 bp of
DNA; 17 Exons.
Transcription
RAF-1 transcribed
nucleotides.
mRNA
contains
3212-3216
Protein
Description
The RAF proteins share three conserved domains: two
(CR1 and CR2) in the N terminus and a third (CR3-
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
239
RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)
Cayo M et al.
RAS (KSR), the multidomain protein connectorenhancer of KSR (CNK), and the leucine-rich-repeat
protein suppressor of RAS mutations-8 (SUR8), which
cause formation of various homo- and heterodimers
and subsequently affect signal transduction. RAF
activation leads to activation of the protein kinases
MEK1 and MEK2 and subsequently the MAPK
proteins ERK1 and ERK2. The downstream effects of
MEK1/2-ERK1/2 activation are varied, complex, and
depend on the cellular context. Resultant effects
include activation of transcription factors involved in
tumorigenesis, cell growth, survival, differentiation,
metabolism, and cytoskeletal rearrangements. RAF-1
(C-RAF-1), A-RAF, and B-RAF are all capable of
activating the MEK1/2-ERK1/2 signaling pathway.
RAF-1 is capable of activating the NF-kB transcription
factor through an unknown mechanism that does not
seem to involve direct phosphorylation of NF-kB and is
independent of MEK1/2-ERK1/2 signaling.
RAF-1 is known to directly affect cell survival through
phosphorylation
of
BAG1
(BCL2-associated
athanogene-1), an anti-apoptotic protein that binds to
BCL2, a second anti-apoptotic factor, also the
prototype for a family of mammalian genes involved in
mitochondrial outer membrane permeability (MOMP),
thus restoring its function. BCL2 also targets RAF-1 to
the mitochondrial membrane, where it is able to more
readily phosphorylate substrates. The RAF1/BAG1/BCL2 interaction allows RAF-1 to
phosphorylate the pro-apoptotic protein BAD at the
mitochondrial membrane, promoting cell survival.
Other known substrates of RAF-1 include the
phosphatase CDC25C, the apoptosis signal-regulating
kinase-1 (ASK1), and the tumor-suppressor protein
retinoblastoma (Rb).
RAF-1 is tightly regulated by the AKT/PKB pathway
through phosphorylation at S259.
of all cases. MTC cells secrete hormones and tumor
markers such as calcitonin, chromogranin A (CgA),
and carcinoembryonic antigen (CEA).
Symptoms are related to either direct invasion or
metastasis (neck mass, dyspnea, dysphagia, voice
changes, pain) or tumor secretion of bioactive amines
and peptides (diarrhea, flushing).
Prognosis
Currently, surgery is the only potentially curative
therapy for patients with MTC. The recommended
operation is total thyroidectomy with lymph node
dissection. However, 50% of patients treated with
surgery suffer persistent or recurrent disease.
Oncogenesis
20% of patients with medullary thyroid cancer have an
autosomal dominant inherited form of the disease,
which is the result of well-characterized point
mutations in the RET proto-oncogene. RAF-1 is
conserved but not expressed at baseline in MTC. Preclinical studies have shown that activation of RAF-1 in
MTC (TT) cells by means of RAF-1 gene transfection
or RAF-1 activating small molecules (ZM336372)
results in tumor cell growth inhibition in vitro and in
vivo.
Carcinoid Tumors
Disease
Carcinoids are tumors that arise from the diffuse
neuroendocrine cell system of the gut, lungs, and other
organs. The incidence is 1-5 per 100,000 individuals.
Carcinoids frequently metastasize to the liver and are
the second most common source of isolated liver
metastases. Carcinoids secrete various bioactive
hormones such as 5-HT (5-hydroxy tryptophan, also
known as serotonin) and chromogranin A.
Prognosis
Patients with hepatic metastases suffer debilitating
symptoms such as abdominal pain, flushing,
bronchoconstriction, and diarrhea. Palliative treatment
for these hormone-induced symptoms includes
somatostatin analogs (such as octeotride). Conventional
anticancer treatments such as chemotherapy and
external beam radiation are largely ineffective for
carcinoid tumors.
Oncogenesis
RAF-1 activation is detrimental to tumorigenesis in
carcinoid cells. Marked reduction in neuroendocrine
phenotypic markers such as human achaete-scute
complex like-1 (ASCL-1) and bioactive hormones 5HT, chromogranin A, and synaptophysin has been
noted upon RAF-1 activation using an estrogeninducible RAF-1 construct in human GI (BON) and
pulmonary carcinoid cell lines (NCI-H727).
Treatment of GI carcinoid cells with RAF-1 activator
ZM336372 led to a decrease in bioactive hormone
levels, a suppression of cellular proliferation, an
Mutations
Somatic
It has been widely established that RAF-1 over activity,
typically via ras-activating mutations, is central to
tumorigenesis and cell proliferation in numerous
cancers (about 30% of all human cancers). However, it
has come to the fore that oncogenesis may be due to
ras/RAF-1 dysregulation (either increased or decreased
expression) rather than increases in ras/RAF-1 activity
exclusively.
Implicated in
Medullary Thyroid Cancer (MTC)
Disease
A neuroendocrine tumor derived from parafollicular C
cells of the thyroid gland, MTC is the third most
common form of thyroid cancer, accounting for 3-5%
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
240
RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)
Cayo M et al.
increase in cell cycle inhibitors p21 and p18, as well as
a decrease in the neuroendocrine phenotypic marker
ASCL-1. ZM336372 treatments also led to progressive
phosphorylation (activation) of MEK1/2, ERK1/2, and
RAF-1.
leads to decreased cell proliferation. RAF-1 activation
in pheochromocytoma cells using ZM336372 led to
cellular differentiation, growth arrest, and a decrease in
the neuroendocrine marker chromogranin A.
Non-Neuroendocrine Cancers with rasactivating Mutations
Small Cell Lung Cancer (SCLC)
Disease
SCLC tends to present with metastatic and regional
spread. Carcinoids rarely metastasize, arise from major
bronchi, and express neuron-specific enolase,
chromogranin, and synaptophysin. Neuroendocrine
carcinoids or atypical carcinoids have a more
aggressive course.
Oncogenesis
Human small-cell lung cancer (SCLC) cell lines rarely
harbor ras-activating mutations. In one cell line of
SCLC, DMS53, it was shown that by RAF-1 induction
using an estrogen-inducible RAF-1 construct SCLC
cells underwent differentiation and G1-specific growth
arrest in conjunction with MEK/ERK1/2 pathway
activation.
Oncogenesis
About 30% of all human cancers express ras-activating
mutations.
More
than
85%
of
pancreatic
adenocarcinomas and 50% of colonic adenocarcinomas
harbor K-ras mutations. K-ras is an upstream effector
of RAF-1 in the RAF-1/MEK/ERK1/2 signaling
pathway. Ras mutations have also been linked to
tumorigenesis of cholangiocarcinoma, adenocarcinoma
of the lung, squamous cell cancer, gastric
adenocarcinoma, small bowel adenocarcinoma, and
malignant melanoma.
Colorectal Cancer
Oncogenesis
RAF-1 is over-activated due to oncogenic ras mutations
in about 50% of colon cancers. These mutations are
associated with poor prognosis, and are necessary for
maintenance of the malignant phenotype.
RAF-1 inhibition in response to interaction with RAF
kinase inhibitor protein (RKIP) (up-regulated in
conjunction with the nuclear factor kappa B signaling
pathway) has been linked with overall and disease-free
survival in patients with colorectal cancers. RKIP has
been identified as potentially useful for identifying
early-stage CRC patients at risk for relapse.
Non-Small Cell Lung Cancer (NSCLC)
Disease
Adenocarcinoma is the most common type of NSCLC
accounting for about 40% of cases. Lesions are
generally located peripherally and develop systemic
metastases despite small primary tumors. 25% of
NSCLC are squamous cell carcinomas which often
remain localized.
Oncogenesis
RAF-1 is over-expressed due to oncogenic ras
mutations in about 35% of NSCLC.
The majority of NSCLC exhibits EGFR overexpression leading to upregulation of RAF-1 activity.
NSCLC has been shown to be mediated by a TGF-a
/EGFR-mediated autocrine loop activated by signaling
involving RAF-1 and PI3K-Akt.
Pancreatic Carcinoma
Oncogenesis
RAF-1 is overactivated due to oncogenic ras mutations
in about 90% of pancreatic carcinomas (Panc-1 and
Mia-PaCa2). It has been shown that malignancy of
these cells is reduced using k-ras RNAi.
Pharmacological inhibition of the RAF/MEK/ERK
pathway in pancreatic cancer cell lines (via MEK
inhibition) results in reduction in cellular proliferation
and an increase in cell cycle arrest.
Pheochromocytoma
Disease
Pheochromocytomas are neuroectodermal in origin and
arise from the chromaffin cells of the adrenal medulla.
10% of tumors are bilateral. Typical symptoms such as
hypertension, headaches, diaphoresis, palpitations,
diarrhea, and skin rashes, are related to tumor
production of catecholamines, especially in patients
with metastases. Pheochromocytoma is potentially
fatal, but relatively uncommon (2-8 cases per million
people annually). Curative therapy is surgery, usually
accomplished by laparoscopic adrenalectomy.
Oncogenesis
Activation of MEK1/2-ERK1/2 is necessary for
differentiation of pheochromocytoma (PC12) cells and
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
Hepatocellular Carcinoma (HCC)
Oncogenesis
RAF-1 is over-activated in about 50% of biopsies while
the RAF-1 protein is over-expressed in nearly 100% of
all HCC's. Angiogenesis and other functions essential
to tumorigenesis in HCC have been reported to depend
on the RAF/MEK/ERK signaling pathway. RAF-1
inhibitor Sorafenib has been reported (in-vitro and invivo) to inhibit RAF-1 activity, leading to decreased
MEK/ERK activity, reduced cellular proliferation, and
apoptosis in several HCC cell lines including HepG2
and PLC/PRF/5.
241
RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)
Cayo M et al.
Prostate Cancer
Renal Cell Carcinoma
Oncogenesis
RAF kinase inhibitor protein (RKIP) coding mRNAs
have been observed to activate interferon-inducible
2',5'-oligoadenylate synthetases (OAS). OAS activity is
characteristically increased (via these mRNAs) in
prostate cancer cell lines PC3, LNCaP and DU145.
RKIP expression is detectable in primary prostate
cancer sections but not in metastases. This suggests
RKIP's characterization as an anti-metastasis gene
using the RAF/MEK/ERK signaling pathway is
appropriate.
RAF-1 inhibition using systemically delivered novel
cationic cardiolipin liposomes (NeoPhectin-AT)
containing a small interfering RNA (siRNA) against
RAF-1 causes tumor growth inhibition in a xenograft
model of human prostate cancer.
RAF/MEK/ERK signaling pathway activation via a
biologically active peptide called a prosaptide (TX14A)
stimulates cell proliferation/survival, migration, and
invasion in human prostate cancer cells.
NSC 95397 and NSC 672121, cdc25 inhibitors, were
shown to activate the RAF/MEK/ERK pathway in
prostate cancer cells.
RAF-1 activation in LNCaP prostate cancer cells using
an estrogen-inducible construct led to growth
inhibition.
Oncogenesis
RAF-1 is overactivated in conjunction with loss of
function of the VHL (von Hippel-Lindau) tumorsuppressor gene.
Breast Cancer
Oncogenesis
RAF-1 inactivation using RNAi in gastric cancer cell
line SGC7901 led to dramatic reductions in
angiogenesis, increased apoptosis, and decreased
cellular proliferation.
Glioma
Oncogenesis
RAF-1 inhibitor AAL881 inhibited growth of glioma
cell xenografts.
Cervical Cancer
Oncogenesis
Low RAF-1 kinase activity is significantly associated
with paclitaxel sensitivity in cervical cancers.
Ovarian Cancer
Oncogenesis
RAF-1 dysregulation is associated with poor prognosis
and possibly carcinogenesis. RAF-1 inhibition using
RNAi reduces cellular proliferatin and inhibits ovarian
tumor cell growth in vitro and in vivo. Similar results
were observed using antisense oligonucleotide (ASO)
therapy (ISIS 5132 and ISIS 13650).
RAF-1 inhibition by the Akt pathway sensitizes human
ovarian cancer cells to the drug paclitaxel.
Gastric Cancer
Oncogenesis
Growth hormone releasing hormone (GHRH) has been
shown to regulate breast cancer cell proliferation and
differentiation. In MDA-231 breast cancer cells,
exogenous
GHRH
stimulated
dose-dependent
proliferation. RAF-1 inhibition using the agent
PD98059 caused prevention of MAPK phosphorylation
by GHRH as well as reduced cellular proliferation.
Proliferative effects of steroid hormone estradiol on
MCF-7 breast cancer cells have been linked with
increased expression of RAF-1, possibly due to direct
activation of RAF-1 by estradiol.
RAF kinase inhibitor protein (RKIP) is associated with
metastasis suppression. RKIP expression is lost in
lymph node metastases. This suggests RKIP is a
metastasis inhibitor gene and that RAF-1 expression
enables metastasis.
The PTK inhibitor AG 879 inhibits proliferation of
human breast cancer cells through inhibition of MAP
kinase activation through inhibition of expression of
the RAF-1 gene.
RAF-1 down-regulation is associated with paclitaxel
drug resistance in human breast cancer cell line MCF7/Adr.
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
Bladder Cancer
Oncogenesis
RAF-1 gene amplification was detected in 4% of
bladder cancer samples. Deletions at the RAF-1 locus
were detected in 2.2% of these samples. Both
amplifications and deletions were heavily correlated
with high tumor grade (P < 0.00001), advanced stage
(P < 0.0001), and poor survival (P < 0.05).
Lymphoma
Oncogenesis
RAF-1 is typically over-expressed
lymphomas from TCR transgenic mice.
in
thymic
References
Bonner TI, Kerby SB, Sutrave P, Gunnell MA, Mark G, Rapp
UR. Structure and biological activity of human homologs of the
raf/mil oncogene. Mol Cell Biol 1985;5(6):1400-1407.
242
RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)
Cayo M et al.
Huebner K, ar-Rushdi A, Griffin CA, Isobe M, Kozak C,
Emanuel BS, Nagarajan L, Cleveland JL, Bonner TI,
Goldsborough MD, et al. Actively transcribed genes in the raf
oncogene group, located on the X chromosome in mouse and
human. Proc Natl Acad Sci USA 1986;83(11):3934-3938.
transformation. Proc Natl Acad Sci USA 2000;97(9):46154620.
Kobzdej M, Matuszyk J, Strzadala L. Overexpression of Ras,
Raf and L-myc but not Bcl-2 family proteins is linked with
resistance to TCR-mediated apoptosis and tumorigenesis in
thymic lymphomas from TCR transgenic mice. Leuk Res
2000;24(1):33-38.
Beck TW, Huleihel M, Gunnell M, Bonner TI, Rapp UR. The
complete coding sequence of the human A-raf-1 oncogene and
transforming activity of a human A-raf carrying retrovirus.
Nucleic Acids Res 1987;15(2):595-609.
Li W, Han M, Guan KL. The leucine-rich repeat protein SUR-8
enhances MAP kinase activation and forms a complex with
Ras and Raf. Genes Dev 2000;14(8):895-900.
Storm SM, Cleveland JL, Rapp UR. Expression of raf family
proto-oncogenes in normal mouse tissues. Oncogene
1990;5(3):345-351.
Luckett JC, Huser MB, Giagtzoglou N, Brown JE, Pritchard CA.
Expression of the A-raf proto-oncogene in the normal adult and
embryonic mouse. Cell Growth Differ 2000;11(3):163-171.
Robbins DJ, Zhen E, Cheng M, Xu S, Ebert D, Cobb MH. MAP
kinases ERK1 and ERK2: pleiotropic enzymes in a ubiquitous
signaling network. Adv Cancer Res 1994;63:93-116.
Chen J, Fujii K, Zhang L, Roberts T, Fu H. Raf-1 promotes cell
survival by antagonizing apoptosis signal-regulating kinase 1
through a MEK-ERK independent mechanism. Proc Natl Acad
Sci USA 2001;98(14):7783-7788.
Barnier J, Papin C, Eychène A, Lecoq O, Calothy G. The
mouse B-raf gene encodes multiple protein isoforms with
tissue-specific expression. The Journal of Biological Chemistry
1995;270(40):23381-23389.
Morrison DK. KSR: a MAPK scaffold of the Ras pathway?. J
Cell Sci 2001;114(Pt 9):1609-1612.
Galaktionov K, Jessus C, Beach D. Raf1 interaction with
Cdc25 phosphatase ties mitogenic signal transduction to cell
cycle activation. Genes Dev 1995;9(9):1046-1058.
Huser M, Luckett J, Chiloeches A, Mercer K, Iwobi M, Giblett
S, Sun XM, Brown J, Marais R, Pritchard C. MEK kinase
activity is not necessary for Raf-1 function. EMBO J
2001;20(8):1940-1951.
Pritchard CA, Bolin L, Slattery R, Murray R, McMahon M. Postnatal lethality and neurological and gastrointestinal defects in
mice with targeted disruption of the A-Raf protein kinase gene.
Curr Biol 1996;6(5):614-617.
McPhillips F, Mullen P, Monia BP, Ritchie AA, Dorr FA, Smyth
JF, Langdon SP. Association of c-Raf expression with survival
and its targeting with antisense oligonucleotides in ovarian
cancer. Br J Cancer 2001;85(11):1753-1758.
Wang HG, Rapp UR, Reed JC. Bcl-2 targets the protein kinase
Raf-1 to mitochondria. Cell 1996;87(4):629-638.
Mikula M,Schreiber M,Husak Z,Kucerova L,Rüth J,Wieser
R,Zatloukal K,Beug H,Wagner EF,Baccarini M. Embryonic
lethality and fetal liver apoptosis in mice lacking the c-raf-1
gene. EMBO J 2001;20(8):1952-1962.
Wang HG, Takayama S, Rapp UR, Reed JC. Bcl-2 interacting
protein, BAG-1, binds to and activates the kinase Raf-1. Proc
Natl Acad Sci USA 1996;93(14):7063-7068.
Wittinghofer A, Nassar N. How Ras-related proteins talk to
their effectors. Trends Biochem Sci 1996;21(12):488-491.
Simon R, Richter J, Wagner U, Fijan A, Bruderer J, Schmid U,
Ackermann D, Maurer R, Alund G, Knönagel H, Rist M, Wilber
K, Anabitarte M, Hering F, Hardmeier T, Schönenberger A,
Flury R, Jäger P, Fehr JL, Schraml P, Moch H, Mihatsch MJ,
Gasser T, Sauter G. High-throughput tissue microarray
analysis of 3p25 (RAF1) and 8p12 (FGFR1) copy number
alterations in urinary bladder cancer. Cancer Res
2001;61(11):4514-4519.
Wojnowski L, Zimmer AM, Beck TW, Hahn H, Bernal R, Rapp
UR, Zimmer A. Endothelial apoptosis in Braf-deficient mice.
Nat Genet 1997;16(3):293-297.
Britten RA, Perdue S, Opoku J, Craighead P. Paclitaxel is
preferentially cytotoxic to human cervical tumor cells with low
Raf-1 kinase activity: implications for paclitaxel-based
chemoradiation regimens. Radiother Oncol 1998;48(3):329334.
Anselmo AN, Bumeister R, Thomas JM, White MA. Critical
contribution of linker proteins to Raf kinase activation. J Biol
Chem 2002;277(8):5940-5943.
Denouel-Galy A, Douville EM, Warne PH, Papin C, Laugier D,
Calothy G, Downward J, Eychene A. Murine Ksr interacts with
MEK and inhibits Ras-induced transformation. Curr Biol
1998;8(1):46-55.
Mabuchi S, Ohmichi M, Kimura A, Hisamoto K, Hayakawa J,
Nishio Y, Adachi K, Takahashi K, Arimoto-Ishida E, Nakatsuji
Y, Tasaka K, Murata Y. Inhibition of phosphorylation of BAD
and Raf-1 by Akt sensitizes human ovarian cancer cells to
paclitaxel. J Biol Chem 2002;277(36):33490-33500.
Pratt MA, Satkunaratnam A, Novosad DM. Estrogen activates
raf-1 kinase and induces expression of Egr-1 in MCF-7 breast
cancer cells. Mol Cell Biochem 1998;189(1-2):119-125.
Pouysségur J, Volmat V, Lenormand P. Fidelity and spatiotemporal control in MAP kinase (ERKs) signalling. Biochem
Pharmacol 2002;64(5-6):755-763.
Wojnowski L, Stancato LF, Zimmer AM, Hahn H, Beck TW,
Larner AC, Rapp UR, Zimmer A. Craf-1 protein kinase is
essential for mouse development. Mech Dev 1998;76(12):141-149.
Sippel RS, Chen H. Activation of the ras/raf-1 signal
transduction pathway in carcinoid tumor cells results in
morphologic transdifferentiation. Surgery 2002;132(6):10351039.
Wang S, Ghosh RN, Chellappan SP. Raf-1 physically interacts
with Rb and regulates its function: a link between mitogenic
signaling and cell cycle regulation. Mol Cell Biol
1998;18(12):7487-798.
Tzivion G, Avruch J. 14-3-3 proteins: active cofactors in cellular
regulation by serine/threonine phosphorylation. J Biol Chem
2002;277(5):3061-3064.
Ravi RK, McMahon M, Yangang Z, Williams JR, Dillehay LE,
Nelkin BD, Mabry M. Raf-1-induced cell cycle arrest in LNCaP
human prostate cancer cells. J Cell Biochem 1999;72(4):45869.
Weinstein-Oppenheimer CR, Burrows C, Steelman LS,
McCubrey JA. The effects of beta-estradiol on Raf activity, cell
cycle progression and growth factor synthesis in the MCF-7
breast cancer cell line. Cancer Biol Ther 2002;1(3):256-262.
Zimmermann S, Moelling K. Phosphorylation and regulation of
Raf by Akt (protein kinase B). Science 1999;286(5445):17411744.
Davis JM, Navolanic PM, Weinstein-Oppenheimer CR,
Steelman LS, Hu W, Konopleva M, Blagosklonny MV,
McCubrey JA. Raf-1 and Bcl-2 induce distinct and common
pathways that contribute to breast cancer drug resistance. Clin
Cancer Res 2003;9(3):1161-1170.
Baumann B, Weber CK, Troppmair J, Whiteside S, Israel A,
Rapp UR, Wirth T. Raf induces NF-kappaB by membrane
shuttle kinase MEKK1, a signaling pathway critical for
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
243
RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1)
Cayo M et al.
Fu Z, Smith PC, Zhang L, Rubin MA, Dunn RL, Yao Z, Keller
ET. Effects of raf kinase inhibitor protein expression on
suppression of prostate cancer metastasis. J Natl Cancer Inst
2003;95(12):878-889.
cardiolipin liposomes silences Raf-1 expression and inhibits
tumor growth in xenograft model of human prostate cancer. Int
J Oncol 2005;26(4):1087-1091.
Van Gompel J.J., Kunnimalaiyaan M, Holen K, Chen H.
ZM336372, a Raf-1 activator, suppresses growth and
neuroendocrine hormone levels in carcinoid tumor cells.
Mol.Cancer.Ther 2005;4, 6:910-917.
Hancock JF. Ras proteins: different signals from different
locations. Nat Rev Mol Cell Biol 2003;4(5):373-384.
Lee M, Koh WS, Han SS. Down-regulation of Raf-1 kinase is
associated with paclitaxel resistance in human breast cancer
MCF-7/Adr cells. Cancer Lett 2003;193(1):57-64.
Al-Mulla F,Hagan S,Behbehani AI,Bitar MS,George SS,Going
JJ,Garcia JJ,Scott L,Fyfe N,Murray GI,Kolch W. Raf kinase
inhibitor protein expression in a survival analysis of colorectal
cancer patients. J Clin Oncol 2006;24(36):5672-5679.
Lanigan TM,Liu A,Huang YZ,Mei L,Margolis B,Guan KL.
Human homologue of Drosophila CNK interacts with Ras
effector proteins Raf and Rlf. FASEB J 2003;17(14):20482060.
Fu Z, Kitagawa Y, Shen R, Shah R, Mehra R, Rhodes D, Keller
PJ, Mizokami A, Dunn R, Chinnaiyan AM, Yao Z, Keller ET.
Metastasis suppressor gene Raf kinase inhibitor protein (RKIP)
is a novel prognostic marker in prostate cancer. Prostate
2006;66(3):248-256.
Morrison DK, Davis RJ. Regulation of MAP kinase signaling
modules by scaffold proteins in mammals. Annu Rev Cell Dev
Biol 2003;19:91-118. (Review).
Gollob JA,Wilhelm S,Carter C,Kelley SL. Role of Raf kinase in
cancer: therapeutic potential of targeting the Raf/MEK/ERK
signal transduction pathway. Semin Oncol 2006;33(4):392-406.
Sippel R.S., Carpenter J.E., Kunnimalaiyaan M., Lagerholm S.,
Chen H. Raf-1 activation suppresses neuroendocrine marker
and hormone levels in human gastrointestinal carcinoid cells.
Am.J.Physiol.Gastrointest.Liver Physiol 2003;285, 2:G245-254.
Kappes A, Vaccaro A, Kunnimalaiyaan M, Chen H. ZM336372,
a Raf-1 activator, inhibits growth of pheochromocytoma cells.
J.Surg.Res 2006;133 (1):42-45.
Troppmair J, Rapp UR. Raf and the road to cell survival: a tale
of bad spells, ring bearers and detours. Biochem Pharmacol
2003;66(8):1341-1345.
Kunnimalaiyaan M, Chen H. The Raf-1 pathway: a molecular
target for treatment of select neuroendocrine tumors?.
Anticancer Drugs 2006;17, 2:139-142.
Keller ET, Fu Z, Yeung K, Brennan M. Raf kinase inhibitor
protein: a prostate cancer metastasis suppressor gene. Cancer
Lett 2004;207(2):131-137.
Letterio J,Rudikoff E,Voong N,Bauer SR. Transforming growth
factor-beta1 sensitivity is altered in Abl-Myc- and Raf-Mycinduced
mouse
pre-B-cell
tumors.
Stem
Cells
2006;24(12):2611-2617.
Koochekpour S, Sartor O, Lee TJ, Zieske A, Patten DY,
Hiraiwa M, Sandhoff K, Remmel N, Minokadeh A. Prosaptide
TX14A stimulates growth, migration, and invasion and
activates the Raf-MEK-ERK-RSK-Elk-1 signaling pathway in
prostate cancer cells. Prostate 2004;61(2):114-123.
Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D,
Wilhelm S, Lynch M, Carter C. Sorafenib blocks the
RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and
induces tumor cell apoptosis in hepatocellular carcinoma
model PLC/PRF/5. Cancer Res 2006;66(24):11851-11858.
Larsson LI. Novel actions of tyrphostin AG 879: inhibition of
RAF-1 and HER-2 expression combined with strong
antitumoral effects on breast cancer cells. Cell Mol Life Sci
2004;61(19-20):2624-2631.
Molinaro RJ, Jha BK, Malathi K, Varambally S, Chinnaiyan AM,
Silverman RH. Selection and cloning of poly(rC)-binding
protein 2 and Raf kinase inhibitor protein RNA activators of
2',5'-oligoadenylate synthetase from prostate cancer cells.
Nucleic Acids Res 2006;34(22):6684-6695.
Mullen P, McPhillips F, MacLeod K, Monia B, Smyth JF,
Langdon SP. Antisense oligonucleotide targeting of Raf-1:
importance of raf-1 mRNA expression levels and raf-1dependent signaling in determining growth response in ovarian
cancer. Clin Cancer Res 2004;10(6):2100-2108.
Mullen P, McPhillips F, Monia BP, Smyth JF, Langdon SP.
Comparison of strategies targeting Raf-1 mRNA in ovarian
cancer. Int J Cancer 2006;118(6):1565-1571.
Nemoto K, Vogt A, Oguri T, Lazo JS. Activation of the Raf1/MEK/Erk kinase pathway by a novel Cdc25 inhibitor in
human prostate cancer cells. Prostate 2004;58(1):95-102.
Sathornsumetee S, Hjelmeland AB, Keir ST, McLendon RE,
Batt D, Ramsey T, Yusuff N,Rasheed BK, Kieran MW, Laforme
A, Bigner DD, Friedman HS, Rich JN. AAL881, a Novel Small
Molecule Inhibitor of RAF and Vascular Endothelial Growth
Factor Receptor Activities, Blocks the Growth of Malignant
Glioma. Cancer Res 2006;66(17):8722-8730.
Wellbrock C, Karasarides M, R Marais. The RAF proteins take
center stage. Nature Reviews Molecular Cell Biology
2004;5(11):875-885.
Chen H, Kunnimalaiyaan M, Van Gompel JJ. Medullary thyroid
cancer: the functions of raf-1 and human achaete-scute
homologue-1. Thyroid 2005;15, 6:511-521.
Siriwardana G,Bradford A,Coy D,Zeitler P. Autocrine/paracrine
regulation of breast cancer cell proliferation by growth hormone
releasing hormone via Ras, Raf, and mitogen-activated protein
kinase. Mol Endocrinol 2006;20(9):2010-2019.
Gysin S, Lee SH, Dean NM, McMahon M. Pharmacologic
inhibition of RAF--->MEK--->ERK signaling elicits pancreatic
cancer cell cycle arrest through induced expression of
p27Kip1. Cancer Res 2005;65(11):4870-4880.
Vaccaro A, Chen H, Kunnimalaiyaan M. In-vivo activation of
Raf-1 inhibits tumor growth and development in a xenograft
model of human medullary thyroid cancer. Anticancer Drugs
2006;17, 7:849-853.
Hagan S, Al-Mulla F, Mallon E, Oien K, Ferrier R, Gusterson B,
García JJ, Kolch W. Reduction of Raf-1 kinase inhibitor protein
expression correlates with breast cancer metastasis. Clin
Cancer Res 2005;11(20):7392-7397.
This article should be referenced as such:
Cayo M, Greentblatt DY, Kunnimalaiyaan M, Chen H. RAF1 (vraf-1 murine leukemia viral oncogene homolog 1). Atlas Genet
Cytogenet Oncol Haematol.2007;11(3):239-244.
Meng F, Ding J, Liu N, Zhang J, Shao X, Shen H, Xue Y, Xie
H, Fan D. Inhibition of gastric cancer angiogenesis by vectorbased RNA interference for Raf-1. Cancer Biol Ther
2005;4(1):113-117.
Pal A, Ahmad A, Khan S, Sakabe I, Zhang C, Kasid UN,
Ahmad I. Systemic delivery of Raf siRNA using cationic
Atlas Genet Cytogenet Oncol Haematol. 2007;11(3)
244