Lithium suppresses astrogliogenesis by neural stem and progenitor cells by inhibiting STAT3 pathway independently of glycogen synthase kinase 3 beta

Zhenzhong Zhu, Penny Kremer, Iman Tadmori, Yi Ren, Dongming Sun, Xijing He, Wise Young, Zhenzhong Zhu, Penny Kremer, Iman Tadmori, Yi Ren, Dongming Sun, Xijing He, Wise Young

Abstract

Transplanted neural stem and progenitor cells (NSCs) produce mostly astrocytes in injured spinal cords. Lithium stimulates neurogenesis by inhibiting GSK3b (glycogen synthetase kinase 3-beta) and increasing WNT/beta catenin. Lithium suppresses astrogliogenesis but the mechanisms were unclear. We cultured NSCs from subventricular zone of neonatal rats and showed that lithium reduced NSC production of astrocytes as well as proliferation of glia restricted progenitor (GRP) cells. Lithium strongly inhibited STAT3 (signal transducer and activator of transcription 3) activation, a messenger system known to promote astrogliogenesis and cancer. Lithium abolished STAT3 activation and astrogliogenesis induced by a STAT3 agonist AICAR (5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside), suggesting that lithium suppresses astrogliogenesis by inhibiting STAT3. GSK3β inhibition either by a specific GSK3β inhibitor SB216763 or overexpression of GID5-6 (GSK3β Interaction Domain aa380 to 404) did not suppress astrogliogenesis and GRP proliferation. GSK3β inhibition also did not suppress STAT3 activation. Together, these results indicate that lithium inhibits astrogliogenesis through non-GSK3β-mediated inhibition of STAT. Lithium may increase efficacy of NSC transplants by increasing neurogenesis and reducing astrogliogenesis. Our results also may explain the strong safety record of lithium treatment of manic depression. Millions of people take high-dose (>1 gram/day) lithium carbonate for a lifetime. GSK3b inhibition increases WNT/beta catenin, associated with colon and other cancers. STAT3 inhibition may reduce risk for cancer.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Heterogeneity of primary NSCs.
Figure 1. Heterogeneity of primary NSCs.
Primary rat NSCs were cultured in growth medium containing 10 ng/ml bFGF and 10 ng/ml EGF for 7 days. A. Neurospheres formed and expressed nestin (A1, green). B. A phase image of adherent NSCs. C–H. Immunostaining of nestin (C, red), GFAP (D, green), Tuj1 (E, red), and GalC (F, red), A2B5 (G, red) and PSA-NCAM (H, green). Nuclei were stained with Hoechst 33342 (DAPI, blue). The table lists percentages of each cell subpopulation relative to total cell count. Data are expressed as mean ± sem averaged from four independent experiments. The scale bar indicates 100 µm.
Figure 2. Inhibition of GSK3β regulates NSC…
Figure 2. Inhibition of GSK3β regulates NSC differentiation.
A. Rat NSCs were cultured in NB27 medium supplemented with LiCl (1 mM), SB216763 (10 µM), or no treatment (control). Cell numbers were estimated with the CyQUANT assay. B. The total cell numbers of untreated (0 mM), LiCl-treated (0.5, 1.0, 3.0, 5.0 mM) and SB216763-treated (10 µM) cultures at 1, 3 and 7 days after passage. C. NSCs were grown for 7 days in NB27 with LiCl (0.5, 1.0, 3.0, 5.0 mM) or SB216763 (10 µM) and then stained for DAPI (blue), Tuj1 (red), and GFAP (green). The photomicrographs (C1) show representative fields from each treatment group (3 mM Lithium, scale bar = 50 µm). The graphs show actual number counts of GFAP+ cells (C2) and Tuj1+ cells (C3), normalized to untreated control counts. D. The expression of GFAP and Tuj1 on NSCs treated with LiCl (0, 0.5, 1.0, 3.0, 5.0 mM) or SB216763 (SB2, 10 µM) after growing 7 days. E1. NSCs were treated with LiCl (0, 0.5, 1.0, 3.0, 5.0 mM) or SB216763 (SB2, 10 µM) for 7 days and apoptotic cells were detected by TUNEL assay (E1, green). Red arrows indicate the typical morphology of apoptotic cells (left two panels, scale bar = 5 µm), the right 6 panels show the staining of TUNEL+ cells in different treatment groups. Nuclei were stained with Hoechst 33342 (blue, scale bar = 25 µm). E2. Percentages of TUNEL+ cells treated with LiCl and SB216763 at indicated time. Data are expressed as mean ± sem from three independent experiments (n = 3, * denotes P<0.05 vs. control, one way ANOVA with Dunnett's post-test).
Figure 3. Inhibition of GSK3β differentially regulates…
Figure 3. Inhibition of GSK3β differentially regulates progenitor cell proliferation.
A1. Immunostaining for A2B5 (green) and Hoechst 33342 (blue). NSCs were grown for 2 days in NB27 media containing LiCl (0, 0.5, 1.0, 3.0, 5.0 mM), or SB216763 (SB2, 10 µM). A2. Quantification of A2B5+ cells as a percentage of total cell count for each LiCl dose and SB216763. B1. The proliferating A2B5+ cells identified by immunostaining for Ki-67 (red) and A2B5 (green). Cells were treated with LiCl or SB216763 for 24 h. B2. Percentages of Ki-67+ cells amongst A2B5+ cells cultured for 24 h in control, 3 mM LiCl, or SB216763-treated NSCs. C1. PSA-NCAM+ (green) co-localized with Tuj1 (red) at this stage, pictures show the percentage of PSA-NCAM+ cells in NSCs treated with LiCl (0, 0.5, 1, 3, 5 mM) or SB216763 (10 µM) for 5 days. C2. The percentage of PSA-NCAM+ cells relative to total cell count. D1. The proliferating PSA-NCAM+ cells identified by immunostaining with Ki-67 (red) and PSA-NCAM (green) after 4 days. D2. The percentage of double Ki-67+/PSA-NCAM+/cells among total PSA-NCAM+ cells. Data are expressed as mean ± sem from three independent experiments, * denotes P<0.05 vs. control (one way ANOVA with Dunnett's post-test).
Figure 4. Lithium suppresses STAT3 activation.
Figure 4. Lithium suppresses STAT3 activation.
A1. LiCl inhibits serum-induced STAT3 activity in a time-dependent manner. Primary rat NSCs were cultured in NB27 medium with 0.5% of FBS in the absence (left) or presence of 3 mM lithium (right) for the indicated time. The STAT3 activity was assessed by detection of phospho-Tyr705-STAT3 (p-STAT3). Similar results were obtained from three independent experiments. B. LiCl inhibits serum-induced STAT3 activity in a dose-dependent manner. NSCs were cultured with various concentrations of LiCl (0.5, 1, 3, 5 mM) in the presence of serum for 24 h. C. Morphological changes of NSCs treated with LiCl, AICAR and LiCl+AICAR, respectively. NSCs received no treatment (Control), lithium (3 mM), AICAR (1 mM), or 45 minutes of lithium pretreatment and addition of AICAR (Li+AICAR). Phase contrast images indicate typical astroglia morphology. D. AICAR induced STAT3 activation and GFAP expression in a time dependent manner. NSCs were treated with 1 mM AICAR for the indicated time and P-STAT3, GFAP and GAPDH were assessed by Western Blot analysis. E. STAT3 (E1) activation and GFAP (E2) expression on NSCs treated with AICAR, lithium or both for 24 hours. F. Expression of Nestin (red) and GFAP (green) on NSCs treated with AICAR, lithium or both for 3 days. G. GFAP expression on NSCs treated with AICAR, lithium or both for 3 days.
Figure 5. Specific GSK3β blockade has no…
Figure 5. Specific GSK3β blockade has no effect on STAT3 activation and astrogliogenesis.
A. Lithium and GSK3β blocker SB216763 inhibit beta catenin phosphorylation (p-beta-Catenin). NSCs were treated with SB216763 (SB2, 10 µM) and LiCl (0, 5, 10, 20 mM) for 30 min. The GSK3β activity was assessed by detection of p-beta-Catenin. B1. SB216763 had no effect on serum-induced STAT3 activation. NSCs were cultured in NB27 medium with 0.5% FBS in the presence of 10 µM SB216763 for the indicated time. B2. Serum increased p-STAT3 over time and SB216763 did not change this curve. Data are expressed as mean ± sem, averaged from three independent experiments and normalized to control values (n = 3, * P<0.05 vs. control, # P<0.05 vs. SB2 treatment group, one way ANOVA with Dunnett's post-test). C. STAT3 activation on NSCs treated with lithium and specific GSK3β inhibitors SB216763 and SB415286. NSCs were treated with LiCl, SB216763, SB415286 and STAT3 inhibitor Stattic at indicated concentrations for 24 h. D. STAT3 activation on NSCs incubated with 1 mM AICAR for 24 h with or without a 45-minute pretreatment of LiCl (3/5 mM) or SB216763 (SB2, 10 µM). E. GFAP expression on NSCs stimulated with AICAR for 3 days in the presence or absence of lithium.
Figure 6. GSK3β inhibition by GID 5-6…
Figure 6. GSK3β inhibition by GID 5-6 does not mimic lithium effect.
NSCs were transfected by electrophoresis with liposomes containing DNA to make Myc-labelled GID5-6, which binds GSK3β and prevents its docking to the cytoplasmic protein axin and phosphorylating beta-catenin. GID5-6/LP is an ineffective analog of GID5-6. A. Transfection efficiency was assessed by immunostaining the cells for Myc (green) after 24 h. Most of the cells were nestin+ (red). B. The effect of GID 5-6 transfection on GSK3β activity. GSK3β activity was assessed by immunoblotting for GSK3β phosphorylated at Ser9 (Ser-P-GSK3β). C. The effect of GID 5-6 transfection on STAT3 activation on NSCs incubated with 0.5% FBS for 24 hours. D. GID5-6 transfection increased cell numbers by 1.2 fold versus GID5-6 LP transfection group as measured by CyQUANT Assay. E. Neither GID5-6 nor GID5-6 LP affected the number of GFAP expressing cells. F. GID5-6 transfection had no effect on number of GFAP-expressing cells. Data were expressed as mean ± sem obtained from three independent experiments (n = 3, * p<0.05 vs. control, t -test). G. GID5-6 transfection did not affect GFAP level on NSCs but lithium (3 mM) markedly reduced GFAP level.

References

    1. Cao QL, Zhang YP, Howard RM, Walters WM, Tsoulfas P, et al. Pluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord are restricted to a glial lineage. Exp Neurol. 2001;167:48–58.
    1. Nakamura M, Okada S, Toyama Y, Okano H. Role of IL-6 in spinal cord injury in a mouse model. Clin Rev Allergy Immunol. 2005;28:197–204.
    1. Mukaino M, Nakamura M, Okada S, Toyama Y, Liu M, et al. [Role of IL-6 in regulation of inflammation and stem cell differentiation in CNS trauma]. Nihon Rinsho Meneki Gakkai Kaishi. 2008;31:93–98.
    1. Lee MY, Kim CJ, Shin SL, Moon SH, Chun MH. Increased ciliary neurotrophic factor expression in reactive astrocytes following spinal cord injury in the rat. Neurosci Lett. 1998;255:79–82.
    1. Tripathi RB, McTigue DM. Chronically increased ciliary neurotrophic factor and fibroblast growth factor-2 expression after spinal contusion in rats. J Comp Neurol. 2008;510:129–144.
    1. Kurek JB, Bennett TM, Bower JJ, Muldoon CM, Austin L. Leukaemia inhibitory factor (LIF) production in a mouse model of spinal trauma. Neurosci Lett. 1998;249:1–4.
    1. Weible MW, 2nd, Chan-Ling T. Phenotypic characterization of neural stem cells from human fetal spinal cord: synergistic effect of LIF and BMP4 to generate astrocytes. Glia. 2007;55:1156–1168.
    1. Ogawa Y, Sawamoto K, Miyata T, Miyao S, Watanabe M, et al. Transplantation of in vitro-expanded fetal neural progenitor cells results in neurogenesis and functional recovery after spinal cord contusion injury in adult rats. J Neurosci Res. 2002;69:925–933.
    1. Okano H, Ogawa Y, Nakamura M, Kaneko S, Iwanami A, et al. Transplantation of neural stem cells into the spinal cord after injury. Semin Cell Dev Biol. 2003;14:191–198.
    1. Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci U S A. 2005;102:14069–14074.
    1. Okada S, Ishii K, Yamane J, Iwanami A, Ikegami T, et al. In vivo imaging of engrafted neural stem cells: its application in evaluating the optimal timing of transplantation for spinal cord injury. Faseb J. 2005;19:1839–1841.
    1. Parr AM, Kulbatski I, Tator CH. Transplantation of adult rat spinal cord stem/progenitor cells for spinal cord injury. J Neurotrauma. 2007;24:835–845.
    1. Sahni V, Kessler JA. Stem cell therapies for spinal cord injury. Nature Reviews Neurology. 2010;6:363–372.
    1. Young W. Review of lithium effects on brain and blood. Cell Transplant. 2009;18:951–975.
    1. Chen G, Rajkowska G, Du F, Seraji-Bozorgzad N, Manji HK. Enhancement of hippocampal neurogenesis by lithium. J Neurochem. 2000;75:1729–1734.
    1. Vazey EM, Connor B. In vitro priming to direct neuronal fate in adult neural progenitor cells. Exp Neurol. 2009;216:520–524.
    1. Lyoo IK, Dager SR, Kim JE, Yoon SJ, Friedman SD, et al. Lithium-induced gray matter volume increase as a neural correlate of treatment response in bipolar disorder: a longitudinal brain imaging study. Neuropsychopharmacology. 2010;35:1743–1750.
    1. Kempton MJ, Geddes JR, Ettinger U, Williams SC, Grasby PM. Meta-analysis, database, and meta-regression of 98 structural imaging studies in bipolar disorder. Arch Gen Psychiatry. 2008;65:1017–1032.
    1. Bearden CE, Thompson PM, Dutton RA, Frey BN, Peluso MA, et al. Three-dimensional mapping of hippocampal anatomy in unmedicated and lithium-treated patients with bipolar disorder. Neuropsychopharmacology. 2008;33:1229–1238.
    1. Sassi RB, Nicoletti M, Brambilla P, Mallinger AG, Frank E, et al. Increased gray matter volume in lithium-treated bipolar disorder patients. Neurosci Lett. 2002;329:243–245.
    1. Su H, Chu TH, Wu W. Lithium enhances proliferation and neuronal differentiation of neural progenitor cells in vitro and after transplantation into the adult rat spinal cord. Exp Neurol. 2007;206:296–307.
    1. Dill J, Wang H, Zhou F, Li S. Inactivation of glycogen synthase kinase 3 promotes axonal growth and recovery in the CNS. J Neurosci. 2008;28:8914–8928.
    1. Yick LW, So KF, Cheung PT, Wu WT. Lithium chloride reinforces the regeneration-promoting effect of chondroitinase ABC on rubrospinal neurons after spinal cord injury. J Neurotrauma. 2004;21:932–943.
    1. Boku S, Nakagawa S, Masuda T, Nishikawa H, Kato A, et al. Glucocorticoids and lithium reciprocally regulate the proliferation of adult dentate gyrus-derived neural precursor cells through GSK-3beta and beta-catenin/TCF pathway. Neuropsychopharmacology. 2009;34:805–815.
    1. Wexler EM, Geschwind DH, Palmer TD. Lithium regulates adult hippocampal progenitor development through canonical Wnt pathway activation. Mol Psychiatry. 2008;13:285–292.
    1. Kim JS, Chang MY, Yu IT, Kim JH, Lee SH, et al. Lithium selectively increases neuronal differentiation of hippocampal neural progenitor cells both in vitro and in vivo. J Neurochem. 2004;89:324–336.
    1. Jope RS. Lithium and GSK-3: one inhibitor, two inhibitory actions, multiple outcomes. Trends Pharmacol Sci. 2003;24:441–443.
    1. Lenox RH, Wang L. Molecular basis of lithium action: integration of lithium-responsive signaling and gene expression networks. Mol Psychiatry. 2003;8:135–144.
    1. Beurel E, Jope RS. Differential regulation of STAT family members by glycogen synthase kinase-3. J Biol Chem. 2008;283:21934–21944.
    1. Bonni A, Sun Y, Nadal-Vicens M, Bhatt A, Frank DA, et al. Regulation of gliogenesis in the central nervous system by the JAK-STAT signaling pathway. Science. 1997;278:477–483.
    1. Lois C, Alvarez-Buylla A. Proliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and glia. Proc Natl Acad Sci U S A. 1993;90:2074–2077.
    1. Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci U S A. 1979;76:4350–4354.
    1. Farr GH, 3rd, Ferkey DM, Yost C, Pierce SB, Weaver C, et al. Interaction among GSK-3, GBP, axin, and APC in Xenopus axis specification. J Cell Biol. 2000;148:691–702.
    1. Zhang F, Phiel CJ, Spece L, Gurvich N, Klein PS. Inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3) in response to lithium. Evidence for autoregulation of GSK-3. J Biol Chem. 2003;278:33067–33077.
    1. Hedgepeth CM, Deardorff MA, Klein PS. Xenopus axin interacts with glycogen synthase kinase-3 beta and is expressed in the anterior midbrain. Mech Dev. 1999;80:147–151.
    1. Hedgepeth CM, Deardorff MA, Rankin K, Klein PS. Regulation of glycogen synthase kinase 3beta and downstream Wnt signaling by axin. Mol Cell Biol. 1999;19:7147–7157.
    1. Lendahl U, Zimmerman LB, McKay RD. CNS stem cells express a new class of intermediate filament protein. Cell. 1990;60:585–595.
    1. Rao MS, Mayer-Proschel M. Glial-restricted precursors are derived from multipotent neuroepithelial stem cells. Dev Biol. 1997;188:48–63.
    1. Mayer-Proschel M, Kalyani AJ, Mujtaba T, Rao MS. Isolation of lineage-restricted neuronal precursors from multipotent neuroepithelial stem cells. Neuron. 1997;19:773–785.
    1. Su H, Zhang W, Guo J, Guo A, Yuan Q, et al. Lithium enhances the neuronal differentiation of neural progenitor cells in vitro and after transplantation into the avulsed ventral horn of adult rats through the secretion of brain-derived neurotrophic factor. J Neurochem. 2009;108:1385–1398.
    1. Fernando P, Brunette S, Megeney LA. Neural stem cell differentiation is dependent upon endogenous caspase 3 activity. FASEB J. 2005;19:1671–1673.
    1. Park DS, So HS, Lee JH, Park HY, Lee YJ, et al. Simvastatin treatment induces morphology alterations and apoptosis in murine cochlear neuronal cells. Acta Otolaryngol. 2009;129:166–174.
    1. Hacker G. The morphology of apoptosis. Cell Tissue Res. 2000;301:5–17.
    1. Lucas FR, Salinas PC. WNT-7a induces axonal remodeling and increases synapsin I levels in cerebellar neurons. Dev Biol. 1997;192:31–44.
    1. Endl E, Gerdes J. The Ki-67 protein: fascinating forms and an unknown function. Exp Cell Res. 2000;257:231–237.
    1. Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U, et al. Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. J Immunol. 1984;133:1710–1715.
    1. Cao F, Hata R, Zhu P, Nakashiro KI, Sakanaka M. Conditional deletion of Stat3 promotes neurogenesis and inhibits astrogliogenesis in neural stem cells. Biochem Biophys Res Commun 2010
    1. Zhu P, Hata R, Cao F, Gu F, Hanakawa Y, et al. Ramified microglial cells promote astrogliogenesis and maintenance of neural stem cells through activation of Stat3 function. FASEB J. 2008;22:3866–3877.
    1. Ni CW, Hsieh HJ, Chao YJ, Wang DL. Shear flow attenuates serum-induced STAT3 activation in endothelial cells. J Biol Chem. 2003;278:19702–19708.
    1. Ma J, Zhang T, Novotny-Diermayr V, Tan AL, Cao X. A novel sequence in the coiled-coil domain of Stat3 essential for its nuclear translocation. J Biol Chem. 2003;278:29252–29260.
    1. Zang Y, Yu LF, Pang T, Fang LP, Feng X, et al. AICAR induces astroglial differentiation of neural stem cells via activating the JAK/STAT3 pathway independently of AMP-activated protein kinase. J Biol Chem. 2008;283:6201–6208.
    1. Hedgepeth CM, Conrad LJ, Zhang J, Huang HC, Lee VM, et al. Activation of the Wnt signaling pathway: a molecular mechanism for lithium action. Dev Biol. 1997;185:82–91.
    1. Orre K, Wennstrom M, Tingstrom A. Chronic lithium treatment decreases NG2 cell proliferation in rat dentate hilus, amygdala and corpus callosum. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:503–510.
    1. Beurel E, Jope RS. Lipopolysaccharide-induced interleukin-6 production is controlled by glycogen synthase kinase-3 and STAT3 in the brain. J Neuroinflammation. 2009;6:9.
    1. Beurel E, Jope RS. Glycogen synthase kinase-3 promotes the synergistic action of interferon-gamma on lipopolysaccharide-induced IL-6 production in RAW264.7 cells. Cell Signal. 2009;21:978–985.
    1. York JD, Ponder JW, Majerus PW. Definition of a metal-dependent/Li(+)-inhibited phosphomonoesterase protein family based upon a conserved three-dimensional core structure. Proc Natl Acad Sci U S A. 1995;92:5149–5153.
    1. Phiel CJ, Klein PS. Molecular targets of lithium action. Annu Rev Pharmacol Toxicol. 2001;41:789–813.
    1. Hallcher LM, Sherman WR. The effects of lithium ion and other agents on the activity of myo-inositol-1-phosphatase from bovine brain. J Biol Chem. 1980;255:10896–10901.
    1. Sherman WR, Gish BG, Honchar MP, Munsell LY. Effects of lithium on phosphoinositide metabolism in vivo. Fed Proc. 1986;45:2639–2646.
    1. Chalecka-Franaszek E, Chuang DM. Lithium activates the serine/threonine kinase Akt-1 and suppresses glutamate-induced inhibition of Akt-1 activity in neurons. Proc Natl Acad Sci U S A. 1999;96:8745–8750.
    1. Ghosh MK, Sharma P, Harbor PC, Rahaman SO, Haque SJ. PI3K-AKT pathway negatively controls EGFR-dependent DNA-binding activity of Stat3 in glioblastoma multiforme cells. Oncogene. 2005;24:7290–7300.
    1. Justicia C, Gabriel C, Planas AM. Activation of the JAK/STAT pathway following transient focal cerebral ischemia: signaling through Jak1 and Stat3 in astrocytes. Glia. 2000;30:253–270.
    1. Acarin L, Gonzalez B, Castellano B. STAT3 and NFkappaB activation precedes glial reactivity in the excitotoxically injured young cortex but not in the corresponding distal thalamic nuclei. J Neuropathol Exp Neurol. 2000;59:151–163.
    1. Cao F, Hata R, Zhu P, Nakashiro K, Sakanaka M. Conditional deletion of Stat3 promotes neurogenesis and inhibits astrogliogenesis in neural stem cells. Biochem Biophys Res Commun. 2010;394:843–847.
    1. Kim OS, Park EJ, Joe EH, Jou I. JAK-STAT signaling mediates gangliosides-induced inflammatory responses in brain microglial cells. J Biol Chem. 2002;277:40594–40601.
    1. Huang C, Ma R, Sun S, Wei G, Fang Y, et al. JAK2-STAT3 signaling pathway mediates thrombin-induced proinflammatory actions of microglia in vitro. J Neuroimmunol. 2008;204:118–125.
    1. Caldwell GM, Jones CE, Soon Y, Warrack R, Morton DG, et al. Reorganisation of Wnt-response pathways in colorectal tumorigenesis. Br J Cancer. 2008;98:1437–1442.
    1. Le Floch N, Rivat C, De Wever O, Bruyneel E, Mareel M, et al. The proinvasive activity of Wnt-2 is mediated through a noncanonical Wnt pathway coupled to GSK-3beta and c-Jun/AP-1 signaling. Faseb J. 2005;19:144–146.
    1. Merdek KD, Nguyen NT, Toksoz D. Distinct activities of the alpha-catenin family, alpha-catulin and alpha-catenin, on beta-catenin-mediated signaling. Mol Cell Biol. 2004;24:2410–2422.
    1. Bordonaro M, Lazarova DL, Carbone R, Sartorelli AC. Modulation of Wnt-specific colon cancer cell kill by butyrate and lithium. Oncol Res. 2004;14:427–438.
    1. Edwards CM, Edwards JR, Lwin ST, Esparza J, Oyajobi BO, et al. Increasing Wnt signaling in the bone marrow microenvironment inhibits the development of myeloma bone disease and reduces tumor burden in bone in vivo. Blood. 2008;111:2833–2842.
    1. Erdal E, Ozturk N, Cagatay T, Eksioglu-Demiralp E, Ozturk M. Lithium-mediated downregulation of PKB/Akt and cyclin E with growth inhibition in hepatocellular carcinoma cells. Int J Cancer. 2005;115:903–910.
    1. Gould TD, Gray NA, Manji HK. Effects of a glycogen synthase kinase-3 inhibitor, lithium, in adenomatous polyposis coli mutant mice. Pharmacol Res. 2003;48:49–53.
    1. He B, You L, Xu Z, Mazieres J, Lee AY, et al. Activity of the suppressor of cytokine signaling-3 promoter in human non-small-cell lung cancer. Clin Lung Cancer. 2004;5:366–370.
    1. He B, You L, Uematsu K, Matsangou M, Xu Z, et al. Cloning and characterization of a functional promoter of the human SOCS-3 gene. Biochem Biophys Res Commun. 2003;301:386–391.
    1. Angelucci F, Aloe L, Jimenez-Vasquez P, Mathe AA. Lithium treatment alters brain concentrations of nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor in a rat model of depression. Int J Neuropsychopharmacol. 2003;6:225–231.
    1. Frey BN, Andreazza AC, Rosa AR, Martins MR, Valvassori SS, et al. Lithium increases nerve growth factor levels in the rat hippocampus in an animal model of mania. Behav Pharmacol. 2006;17:311–318.
    1. Machado-Vieira R, Manji HK, Zarate CA., Jr The role of lithium in the treatment of bipolar disorder: convergent evidence for neurotrophic effects as a unifying hypothesis. Bipolar Disord. 2009;11(Suppl 2):92–109.
    1. Walz JC, Frey BN, Andreazza AC, Cereser KM, Cacilhas AA, et al. Effects of lithium and valproate on serum and hippocampal neurotrophin-3 levels in an animal model of mania. J Psychiatr Res. 2008;42:416–421.

Source: PubMed

3
購読する