You are on page 1of 5

Neuroscience Letters 444 (2008) 92–96

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Raf inhibition protects cortical cells against ␤-amyloid toxicity


Valentina Echeverria a,b,∗ , Sarah Burgess a , Joyonna Gamble-George a,b ,
Gary W. Arendash c,d,e , Bruce A. Citron a,b
a
Bay Pines VA Healthcare System, Bay Pines, FL 33744, USA
b
Department of Molecular Medicine, University of South Florida, Tampa, FL 33647, USA
c
The Johnnie B. Byrd, Sr. Alzheimer’s Center and Research Institute, Tampa, FL 33613, USA
d
Florida Alzheimer’s Disease Research Center, University of South Florida, Tampa, FL 33612, USA
e
Division of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA

a r t i c l e i n f o a b s t r a c t

Article history: Alzheimer’s disease (AD) is the main cause of dementia in the elderly. The discovery of new targets of
Received 16 April 2008 therapeutic intervention is fundamental to the development of new drugs against AD pathology. Upreg-
Received in revised form 23 July 2008 ulation of cRaf-1 has been found post-mortem in the brains of AD patients. cRaf-1 is a cytosolic protein
Accepted 25 July 2008
kinase that regulates neuronal survival and senescence. In this study, we investigated cRaf-1 in the brains
of aged APPswe mice presenting AD-like pathology and whether Raf inhibitors protected cultured cortical
Keywords:
cells against amyloid ␤ toxicity (A␤). We found a dysregulation of cRaf-1 in the cortex of APPswe mice,
Alzheimer’s
which showed a 147% increase in the active form phosphorylated at serine 338 and a 40% decrease in the
Neuroinflammation
Neuroprotection
levels of the inactive form of cRaf-1, phospho-cRaf-1[Ser259]. Furthermore, treatment of primary cortical
SN50 neurons with the cRaf-1 inhibitors, GW5074 or ZM336372, and the nuclear factor kappa B (NF␬B) inhibitor
SN50, protected cortical neurons against A␤ toxicity. Since Raf stimulates NF␬B, we studied the effect of
Raf inhibition on its activation by studying changes in NF␬B phosphorylation at serine 276. Our results
suggest that Raf inhibition with GW5074 is neuroprotective against A␤ toxicity through a mechanism that
involves NF␬B inhibition.
© 2008 Elsevier Ireland Ltd. All rights reserved.

A great deal of published evidence supports the concept that To investigate whether cRaf-1 dysregulation is a general mecha-
the accumulation of amyloid ␤-peptide (A␤) in the brain induces nism of AD, we analyzed cRaf-1 expression and phosphorylation in
synaptic dysfunction and neuronal cell death. The use of cultured the brains of aged APPswe mice. Since Raf overactivation is a molec-
neurons and transgenic AD animal models such as the APPswe mice ular hallmark in brains exposed to high levels of A␤, we investigated
has permitted the discovery of several molecular changes induced the effect of the Raf inhibitors GW5074 and ZM336372 on A␤ tox-
by A␤ toxicity and potential therapeutic targets against AD. icity in cultured cortical cells. A previous study showed that these
The neurochemical analyses of the brain of patients with AD cRaf-1 inhibitors protected cerebellar neurons against other neuro-
showed that high levels of A␤ are accompanied by changes in toxins by a mechanism that required NF␬B activity [6]. In the same
several protein kinases involved in neuronal survival such as cAMP- report, acute treatment with GW5074 decreased neurodegenera-
dependent protein kinase A (PKA) [23], extracellular regulated tion in a mouse model of Huntington’s disease [6]. cRaf-1 stimulates
kinase (ERK) [10], and cRaf-1 [26]. cRaf-1 activity is controlled NF␬B activity by activating the regulator inhibitor ␬B (I␬B) kinase
by phosphorylation at two sites; cRaf-1 is generally found to be (IKK) [21]. When activated, IKK induces the degradation of the
inactive when phosphorylated at serine 259, and active when phos- inhibitor of NF␬B, I␬B␣. Then, NF␬B translocates into the nucleus
phorylated at serine 338 [2,18]. The dysregulation of cRaf-1 found where it stimulates the expression of amyloid ␤-precursor protein
in AD brains is coherent with a decrease in proteins that inhibit its (APP) and ␤-secretase 1 (BACE1) [3]. The Raf inhibitor GW5074
activity, such as the Raf kinase inhibitor protein (RKIP) [11] and PKA, decreased the phosphorylation of NF␬B at the activation site serine
[22] and an increase of proteins that stimulate PKA such as Ras [25]. 276, suggesting that GW5074 is neuroprotective against A␤ toxicity
by inhibiting NF␬B.
Most chemicals including GW5074 [6] and ZM336372 were
∗ Corresponding author at: Bay Pines VA Healthcare System, 10,000 Bay Pines
purchased from Sigma Chemicals (St. Louis, MO, USA) and all anti-
Blvd., Building 23, Room 123, Bay Pines, FL 33744, USA. Tel.: +1 727 398 6661x4425;
bodies were purchased from Cell Signaling, Inc. (Beverly, MA, USA)
fax: +1 727 319 1161. unless specified otherwise. SN50 was purchased from Calbiochem
E-mail address: valentina.echeverria@va.gov (V. Echeverria). (San Diego, CA, USA). A␤1–42 peptide was obtained from American

0304-3940/$ – see front matter © 2008 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.neulet.2008.07.092
V. Echeverria et al. / Neuroscience Letters 444 (2008) 92–96 93

Peptide (Sunnyvale, CA, USA). Hoechst 33342, propidium iodide, 0.05% Tween-20 (PBST) containing 10% skim milk. Membranes
dichlorodihydrofluorescein diacetate (CM-H2 DCFDA) and materi- were incubated with primary antibodies in PBST with 3% dry milk
als used for cell culture were obtained from Invitrogen (Carlsbad, overnight at 4 ◦ C and with secondary antibodies for 2 h. Rabbit poly-
CA, USA). clonal antibodies were directed against pcRaf-1 (Ser259) (1:500)
The in vivo studies were performed using 16–18-month- and pcRaf-1 (Ser338) (1:500) (Cell Signaling, Inc., CA, USA), and
old transgenic APPswe mice containing the Swedish mutation pNF␬B/p65 (Ser276) (1:500) (GenScript Corporation, NJ, USA). A
(K260N/M671L) and age-matched wild-type littermate mice. monoclonal mouse antibody, anti ␤-tubulin (Promega, WI, USA),
Mouse brains were removed after euthanasia by cervical disloca- was used as a control for protein loading. The bands were detected
tion, and the cortex dissected out and frozen at −80 ◦ C until use. using ECL detection kit (ECL, Pharmacia Biotech, Piscataway, NJ,
Mice were maintained on a 12 h dark and 12 h light cycle with USA), visualized using the Kodak Image Station 440CF and analyzed
ad libitum access to food and water. Animals were used in accor- using the NIH ImageJ software. All data was normalized against
dance with the National Institutes of Health guidelines for the use tubulin immunoreactivity and expressed as percentage of control
of experimental animals. Protocols were approved by the Institu- values.
tional Animal Care and Use Committee of the University of South We and others have found that the active form of cRaf-1 is upreg-
Florida, and Bay Pines and Tampa VA Healthcare Systems. ulated in the cortex of human AD brains [26]. To investigate whether
Embryonic rat cortical neurons were cultured following the pro- cRaf-1 dysregulation was a common characteristic of AD brains, we
tocol described previously [4] with minor modifications. Briefly, analyzed by Western blot the levels of the inactive form phospho-
cerebral cortices were obtained from BrainBits LLC (Springfield, IL, rylated at serine 259 and the active form phosphorylated at serine
USA). The brain tissues were dissociated by 0.05% (v/v) trypsin 338 of cRaf-1 in the cortex of APPswe mice. We found a signifi-
digestion and triturated. Cells (1250 cells/mm2 ) were plated in cant decrease in the inactive form of cRaf-1 (mean ± S.E.M.: 63 ± 6%;
Neurobasal medium E, supplemented with 1 mM glutamax, and P = 0.02, n = 3) (37% reduction) (Fig. 1A) and a significant increase in
2% B27 and plated onto 24-well plates coated with poly-d-lysine the levels of the active form (mean ± S.E.M.: 247 ± 56%; P = 0.038,
(0.1 mg/ml). The cell cultures were kept at 37 ◦ C in a humidified n = 4) (147% increase) in the cortex of 16–18-month-old APPswe
incubator with 95% air/5%CO2 until used. mice compared to wild-type control littermates (mean ± S.E.M.:
After 7–10 days in vitro (DIV), the media was removed and 100 ± 26%; n = 5) (Fig. 1B). The characteristic double bands migrated
the cortical cells were incubated with Neurobasal medium sup- with a relative molecular weight of 75–100 kDa.
plemented with B27 minus antioxidant (B27-AO; Invitrogen) for To investigate the effect of cRaf-1 inhibitors on neuronal survival
2 h. Then cells were exposed to A␤1–42 oligomers in the presence after A␤1–42 toxicity, cortical cells were co-treated with A␤ peptide
or absence of GW5074 or ZM336372. A␤1–42 solution was pre- 5 ␮M and different concentrations of either GW5074 or ZM336372,
pared to obtain oligomers by dissolving 0.5 mg of A␤1–42 peptide and after 48 h cell viability was analyzed. The co-treatment of neu-
in a solution of sodium hydroxide (1 mM) and an equal volume of rons with both Raf inhibitors was protective against A␤1–42 toxicity.
phosphate-buffered saline (PBS), pH 7.4 (Invitrogen). The A␤1–42 The co-treatment of cells with the Raf inhibitors increased cell sur-
solution was then diluted into the culture medium to reach a final vival from 73% (5 ␮M A␤) to 93% (5 ␮M A␤ + GW5074, 10 ␮M) and
concentration of 5 ␮M as previously described [8]. All assays were from 70% (5 ␮M A␤) to 86% (5 ␮M A␤ + ZM336372 10 ␮M) of control
performed in triplicate and repeated at least three times. levels as estimated by MTT assay (Fig. 2A and B). A similar pattern
Cell viability was quantified by the MTT assay that mea- of neuroprotection was observed using calcein-AM (live cells) and
sures the mitochondrial conversion of the tetrazolium salt MTT PI (dying cells) staining. The number of PI stained cells after A␤
(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) exposure decreased dramatically when cells were co-treated with
to formazan as described [4]. Cells were incubated with MTT 10 ␮M GW5074 (Fig. 2C).
(0.5 mg/ml) for 1–3 h at 37 ◦ C; MTT formazan crystals were dis- It has been found that NF␬B/p65 expression is elevated in brain
solved in DMSO and absorbance at 570 nm was measured. Cell cells surrounding senile plaques in the brains accumulating A␤ [15].
viability was also estimated with propidium iodide (PI) and cal- Moreover, the inhibition of NF␬B is protective against A␤ neuro-
cein. 7 DIV cortical cells were incubated in 1 ml of media containing toxicity [5]. To investigate whether NF␬B activity was involved in
1 ␮g/ml of PI and calcein-AM for 30 min at 37 ◦ C. After incuba- the beneficial actions of GW5074, we studied the effect of SN50 on
tion, cells were washed with PBS and fluorescence was determined GW5074 activity. We found that the treatment of neurons with the
with a fluorescence microscope (Microscope Leica DMI 4000B, 20×, NF␬B inhibitor SN50, alone or in the presence of 10 ␮M GW5074,
40×). The changes in cell viability were determined by analyzing induced similar levels of neuroprotection against A␤ toxicity as
the number of calcein-AM (green, living) and PI (red nucleus, dead) GW5074 (Fig. 3A).
cells in any experimental condition. At least 300 cells were analyzed To determine the effect of GW5074 on the A␤-induced activa-
by condition. Assays were measured in triplicate and repeated at tion of NF␬B, we investigated NF␬B/p65 phosphorylation at serine
least twice. Results are expressed as percentage of vehicle-treated 276 in cortical cells treated with: (a) PBS; (b) 5 ␮M A␤; and (c)
controls. 5 ␮M A␤ plus 10 ␮M GW5074. We found that cells treated with
Cortical cells (0.5 × 106 cells/well) after 7 DIV and brain tissues 5 ␮M A␤ showed an almost three-fold increase in NF␬B/p65 phos-
from 16–18-month-old APPswe mice were disrupted by sonication phorylation at serine 276 (mean ± S.E.M.: 270 ± 65%) compared to
in cold lysis buffer (Cell Signaling Technology, Beverly, MA, USA) vehicle-treated control cells (mean ± S.E.M.: 100 ± 14%). Cells co-
containing a complete protease inhibitor cocktail (Roche Molecular treated with 10 ␮M GW5074 and 5 ␮M A␤ presented a nine-fold
Biochemicals, Indianapolis, IN, USA), 1 mM phenylmethyl sulpho- decrease in NF␬B/p65 phosphorylation in comparison to A␤ treated
nyl fluoride (PMSF; Sigma, Saint Louis, MI, USA), and phosphatase cells (Fig. 3B).
inhibitors (Sigma). After sonication, protein extracts were incu- We investigated cRaf-1 expression and activity in the cortex
bated on ice and centrifuged to 16,000 × g for 30 min at 4 ◦ C and of APPswe mice and the effect of the cRaf-1 inhibitors GW5074
supernatants were used for Western blot analysis. and ZM336372 on A␤-dependent activation of NF␬B and neurotox-
Equal amounts of protein were separated by sodium dode- icity. The cRaf-1 inhibitors provided neuroprotection against A␤
cyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and toxicity through a mechanism that was mimicked by the NF␬B
transferred to nitrocellulose membranes (BA83 0.2 ␮m; Bio-Rad). inhibitor SN50. Supporting the idea that Raf inhibitors are neu-
The membranes were blocked in phosphate buffered saline with roprotective by inhibiting NF␬B, we found that GW5074 inhibited
94 V. Echeverria et al. / Neuroscience Letters 444 (2008) 92–96

Fig. 1. Dysregulation of cRaf-1 in the cortex of APPswe mice. We analyzed cRaf-1 phosphorylation in the cortex of 16–18-month-old APPswe mice and age-matched wild-
type control littermates (WT). Cortical tissues were homogenized in lysis buffer containing protease and phosphatase inhibitors, and the immunoreactivity for cRaf-1
phosphorylated at serines 338 and 259 was determined by Western blotting. Values were calculated as percentage of wild-type mice levels considered as 100% and expressed
as mean ± S.E.M. * Significant difference of the means as analyzed using student t-test.

the A␤-dependent activation of NF␬B. Since we obtained neuropro- hallmark of AD and a key factor inducing A␤ synthesis. One of the
tection using two different cRaf-1 inhibitors, we postulate that Raf obvious explanations of the abnormal activation of cRaf-1 in the
inhibition underlies its beneficial actions. Coherent with the inhibi- brain of AD patients is the increased activity of the upstream activa-
tion of cRaf-1 by GW5074, we observed a decrease in the activation tor of cRaf-1, Ras [9]. It has been hypothesized that Ras can promote
by phosphorylation of NF␬B at serine 276 in cultured cortical neu- apoptosis in the brain by triggering the re-entrance of neurons into
rons. Our results suggest that cRaf-1 hyperactivity is a molecular the cell cycle [10].

Fig. 2. Raf-1 inhibitors are neuroprotective against A␤ toxicity. Embryonic rat cortical cells were cultured in Neurobasal/B27 media. After 7 DIV, cells were co-treated
with 5 ␮M A␤ and different concentrations of the Raf inhibitors, GW5074 (A and C) and ZM336372 (B). After 48 h, cell viability was analyzed by using MTT (A and B) and
calcein/propidium iodide (PI) assays (C). Cell viability values were normalized as percentage of the control cell values considered as 100% and expressed as mean ± S.E.M.
***
Significant difference P < 0.001. ** P < 0.01. ns, No significant difference. A␤, Amyloid beta peptide.
V. Echeverria et al. / Neuroscience Letters 444 (2008) 92–96 95

Fig. 3. GW5074 protects cortical cells against A␤ toxicity by inhibiting NF␬B. Cortical cells were cultured in Neurobasal/B27 media. After 7 DIV, cells were treated with 5 ␮M
A␤ alone or co-treated with the Raf inhibitor GW5074 and/or the NF␬B inhibitor SN50. After 48 h, cell viability was analyzed by using MTT and expressed as percentage of the
controls (A). To investigate the effect of GW5074 on NF␬B phosphorylation at serine 276, after 7 DIV cortical cells were treated with vehicle (control), 5 ␮M A␤ and 5 ␮M A␤
plus 10 ␮M GW5074 for 48 h. After treatment, cells were homogenized in lysis buffer containing protease and phosphatase inhibitors, and equal amounts of protein samples
(60 ␮g) were analyzed by Western blot (B). The cell viability results were normalized to control values considered as 100% and expressed as mean ± S.E.M. *** Significant
difference P < 0.001. ns, No significant difference.

Moreover, abnormal cRaf-1 activation can be due to the decrease tered orally to the patients. We are currently investigating the use
of endogenous inhibitors such as PKA [17] and the Raf kinase of sorafenib in vivo against AD.
inhibitor protein (RKIP) [16]. Since PKA activity is downregulated
in AD brains, this may be another factor causing cRaf-1 dysreg- Acknowledgments
ulation. In relation to the mechanism underlying the beneficial
effects of Raf inhibitors, we found that the protection conferred We thank Steve Dennis and Ross Zeitlin for technical and edi-
by GW5074 against A␤ paralleled a decrease of NF␬B/p65 activa- torial assistance. This work was supported by the Johnnie B. Byrd,
tion. It is well known that after its activation by phosphorylation at Sr. Alzheimer’s Center and Research Institute pilot grant (to V.E.
serine 338 [7], cRaf-1 stimulates NF␬B activity [24]. NF␬B/p65, one and B.C.), funds from the Florida Alzheimer’s Disease Research Cen-
of the more abundant NF␬B subunits in the central nervous sys- ter (to G.A.), and the James and Esther King Biomedical Research
tem, is upregulated in the brains of patients suffering from several New Investigator Program grant (to V.E.). We are also grateful to
neurodegenerative conditions including Parkinson’s disease, Amy- the Alzheimer’s Association, VA Medical Research Service, The Bay
otrophic lateral sclerosis [1,12] and AD [14]. It is feasible that the Pines Foundation and Dr. Hugo Fernandez for constant support and
inhibition of NF␬B activation by Raf inhibitors can be also benefi- encouragement.
cial in these conditions. Such a mechanism would be in line with
numerous reports showing that inhibition of NF␬B reduces A␤ tox- References
icity [30] and its production [28].
NF␬B is inhibited by many neuroprotective compounds against [1] S.W. Barger, A.M. Moerman, X. Mao, Molecular mechanisms of cytokine-
induced neuroprotection: NFkappaB and neuroplasticity, Curr. Pharm. Des. 11
AD [27] such as resveratrol, quercetin, Ginkgo Biloba and curcumin.
(2005) 985–998.
We propose that a similar mechanism is underlying the observed [2] M. Beeram, A. Patnaik, E.K. Rowinsky, Regulation of c-Raf-1: therapeutic impli-
neuroprotective activity of Raf inhibitors against A␤ toxicity. For cations, Clin. Adv. Hematol. Oncol. 1 (2003) 476–481.
example, the actions of quercetin against cancer are attributed to [3] K.Z. Bourne, D.C. Ferrari, C. Lange-Dohna, S. Rossner, T.G. Wood, J.R. Perez-Polo,
Differential regulation of BACE1 promoter activity by nuclear factor-kappaB in
the inhibition of Raf and the resultant decrease in NF␬B activity neurons and glia upon exposure to beta-amyloid peptides, J. Neurosci. Res. 85
[20]. The expected outcome of NF␬B inhibition includes a decrease (2007) 1194–1204.
in the expression of the A␤ synthesizing enzymes such as BACE1 [4] G.J. Brewer, Serum-free B27/neurobasal medium supports differentiated
growth of neurons from the striatum, substantia nigra, septum, cerebral cortex,
[3]. Several Raf inhibitors have been developed for the treatment of cerebellum, and dentate gyrus, J. Neurosci. Res. 42 (1995) 674–683.
cancer. One of these inhibitors, sorafenib, which is already approved [5] J. Chen, Y. Zhou, S. Mueller-Steiner, L.F. Chen, H. Kwon, S. Yi, L. Mucke, L. Gan,
by the Food and Drug Administration as a treatment for kidney SIRT1 protects against microglia-dependent amyloid-beta toxicity through
inhibiting NF-kappaB signaling, J. Biol. Chem. 280 (2005) 40364–40374.
[13] and liver cancers [19], is particularly interesting because of its [6] P.C. Chin, L. Liu, B.E. Morrison, A. Siddiq, R.R. Ratan, T. Bottiglieri, S.R. D’Mello,
minimal side effects in humans [29] and its ability to be adminis- The c-Raf inhibitor GW5074 provides neuroprotection in vitro and in an ani-
96 V. Echeverria et al. / Neuroscience Letters 444 (2008) 92–96

mal model of neurodegeneration through a MEK-ERK and Akt-independent [18] M. Kunnimalaiyaan, H. Chen, The Raf-1 pathway: a molecular target for treat-
mechanism, J. Neurochem. 90 (2004) 595–608. ment of select neuroendocrine tumors? Anticancer Drugs 17 (2006) 139–142.
[7] A.S. Dhillon, A. von Kriegsheim, J. Grindlay, W. Kolch, Phosphatase and feedback [19] L. Lang, FDA approves sorafenib for patients with inoperable liver cancer, Gas-
regulation of Raf-1 signaling, Cell Cycle 6 (2007) 3–7. troenterology 134 (2008) 379.
[8] V. Echeverria, A. Clerman, S. Dore, Stimulation of PGE receptors EP2 and EP4 [20] K.W. Lee, N.J. Kang, Y.S. Heo, E.A. Rogozin, A. Pugliese, M.K. Hwang, G.T. Bowden,
protects cultured neurons against oxidative stress and cell death following A.M. Bode, H.J. Lee, Z. Dong, Raf and MEK protein kinases are direct molecular
beta-amyloid exposure, Eur. J. Neurosci. 22 (2005) 2199–2206. targets for the chemopreventive effect of quercetin, a major flavonol in red
[9] I. Ferrer, R. Blanco, M. Carmona, B. Puig, Phosphorylated c-MYC expression in wine, Cancer Res. 68 (2008) 946–955.
Alzheimer disease, Pick’s disease, progressive supranuclear palsy and corti- [21] S. Li, J.M. Sedivy, Raf-1 protein kinase activates the NF-kappa B transcription
cobasal degeneration, Neuropathol. Appl. Neurobiol. 27 (2001) 343–351. factor by dissociating the cytoplasmic NF-kappa B-I kappa B complex, Proc.
[10] I. Ferrer, R. Blanco, M. Carmona, R. Ribera, E. Goutan, B. Puig, M.J. Rey, A. Car- Natl. Acad. Sci. U.S.A. 90 (1993) 9247–9251.
dozo, F. Vinals, T. Ribalta, Phosphorylated map kinase (ERK1, ERK2) expression [22] Y.Q. Liang, X.T. Huang, X.C. Tang, Huperzine A reverses cholinergic and
is associated with early tau deposition in neurones and glial cells, but not with monoaminergic dysfunction induced by bilateral nucleus basalis magnocel-
increased nuclear DNA vulnerability and cell death, in Alzheimer disease, Pick’s lularis injection of beta-amyloid peptide (1–40) in rats, Cell. Mol. Neurobiol.
disease, progressive supranuclear palsy and corticobasal degeneration, Brain (2007).
Pathol. (Zurich, Switzerland) 11 (2001) 144–158. [23] Z. Liang, F. Liu, I. Grundke-Iqbal, K. Iqbal, C.X. Gong, Down-regulation of cAMP-
[11] A.J. George, R.M. Holsinger, C.A. McLean, S.S. Tan, H.S. Scott, T. Cardamone, R. dependent protein kinase by over-activated calpain in Alzheimer disease brain,
Cappai, C.L. Masters, Q.X. Li, Decreased phosphatidylethanolamine binding pro- J. Neurochem. 103 (2007) 2462–2470.
tein expression correlates with Abeta accumulation in the Tg2576 mouse model [24] Q. Liu, J. Fan, M. McMahon, A.M. Prince, P. Zhang, Role of the oncogenic Raf-1 in
of Alzheimer’s disease, Neurobiol. Aging 27 (2006) 614–623. orchestration of discrete nuclear factor-kappaB-activating pathways, Mol. Cell.
[12] A. Ghosh, A. Roy, X. Liu, J.H. Kordower, E.J. Mufson, D.M. Hartley, S. Ghosh, R.L. Biol. Res. Commun. 4 (2001) 381–389.
Mosley, H.E. Gendelman, K. Pahan, Selective inhibition of NF-kappaB activation [25] A. McShea, D.A. Zelasko, J.L. Gerst, M.A. Smith, Signal transduction abnormal-
prevents dopaminergic neuronal loss in a mouse model of Parkinson’s disease, ities in Alzheimer’s disease: evidence of a pathogenic stimuli, Brain Res. 815
Proc. Natl. Acad. Sci. U.S.A. 104 (2007) 18754–18759. (1999) 237–242.
[13] T.E. Hutson, Safety and tolerability of sorafenib in clear-cell renal cell [26] M. Mei, B. Su, K. Harrison, M. Chao, S.L. Siedlak, L.A. Previll, L. Jackson, D.X. Cai, X.
carcinoma: a Phase III overview, Exp. Rev. Anticancer Therapy 7 (2007) Zhu, Distribution, levels and phosphorylation of Raf-1 in Alzheimer’s disease,
1193–1202. J. Neurochem. 99 (2006) 1377–1388.
[14] B. Kaltschmidt, M. Uherek, B. Volk, P.A. Baeuerle, C. Kaltschmidt, Transcription [27] N.H. Nam, Naturally occurring NF-kappaB inhibitors, Mini Rev. Med. Chem. 6
factor NF-kappaB is activated in primary neurons by amyloid beta peptides and (2006) 945–951.
in neurons surrounding early plaques from patients with Alzheimer disease, [28] D. Paris, N. Patel, A. Quadros, M. Linan, P. Bakshi, G. Ait-Ghezala, M. Mullan, Inhi-
Proc. Natl. Acad. Sci. U.S.A. 94 (1997) 2642–2647. bition of Abeta production by NF-kappaB inhibitors, Neurosci. Lett. 415 (2007)
[15] B. Kaltschmidt, M. Uherek, H. Wellmann, B. Volk, C. Kaltschmidt, Inhibition of 11–16.
NF-kappaB potentiates amyloid beta-mediated neuronal apoptosis, Proc. Natl. [29] C.H. Takimoto, A. Awada, Safety and anti-tumor activity of sorafenib (Nexavar)
Acad. Sci. U.S.A. 96 (1999) 9409–9414. in combination with other anti-cancer agents: a review of clinical trials, Cancer
[16] E.T. Keller, Z. Fu, M. Brennan, The role of Raf kinase inhibitor protein (RKIP) in Chemother. Pharmacol. 61 (2008) 535–548.
health and disease, Biochem. Pharmacol. 68 (2004) 1049–1053. [30] A. Valerio, F. Boroni, M. Benarese, I. Sarnico, V. Ghisi, L.G. Bresciani, M. Fer-
[17] S.H. Kim, A.C. Nairn, N. Cairns, G. Lubec, Decreased levels of ARPP-19 and PKA rario, G. Borsani, P. Spano, M. Pizzi, NF-kappaB pathway: a target for preventing
in brains of Down syndrome and Alzheimer’s disease, J. Neural Transm. Suppl. beta-amyloid (Abeta)-induced neuronal damage and Abeta42 production, Eur.
(2001) 263–272. J. Neurosci. 23 (2006) 1711–1720.

You might also like