Articles

  • KSBNS 2024

Article

Review Article

Exp Neurobiol 2013; 22(3): 167-172

Published online September 30, 2013

https://doi.org/10.5607/en.2013.22.3.167

© The Korean Society for Brain and Neural Sciences

Promise of Neurorestoration and Mitochondrial Biogenesis in Parkinson's Disease with Multi Target Drugs: An Alternative to Stem Cell Therapy

Moussa BH Youdim1* and Young J. Oh2

1Abital Pharma Pipeline Ltd, 96 Yuval Alon St., 61500 Tel Aviv, Israel, 2Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 120-749, Korea

Correspondence to: *To whom correspondence should be addressed.
TEL: 972-4-9090000, FAX: 972-4-9090001
e-mail: youdim@tx.technion.ac.il

There is an unmet need in progressive neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. The present therapeutics for these diseases at best is symptomatic and is not able to delay disease or possess disease modifying activity. Thus an approach to drug design should be made to slow or halt progressive course of a neurological disorder by interfering with a disease-specific pathogenetic process. This would entail the ability of the drug to protect neurons by blocking the common pathway for neuronal injury and cell death and the ability to promote regeneration of neurons and restoration of neuronal function. We have now developed a number of multi target drugs which possess neuroprotective, and neurorestorative activity as well as being able to active PGC-1α (peroxisome proliferator-activated receptor γ coactivator-1α), SIRT1 (NAD-dependent deacetylase protein) and NTF (mitochondrial transcription factor) that are intimately associated with mitochondrial biogenesis.

Keywords: Parkinson's disease, neuroprotective, neurorestorative, multi target drug, iron chelator, mitochondrial biogenesis

There are significant evidence for dysregulation of brain iron metabolism in neurodegenerative disease of Parkinson's disease (PD), Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Iron is thought to participate in oxidative stress initiated by the Fenton reaction [1] and monoamine oxidase generating hydrogen peroxide. Thus, the concept of iron chelation as a valuable therapeutic approach in neurological disorders led our group to develop multi target, nontoxic, lipophilic, brain permeable compounds with iron chelating-radical scavenging, monoamine oxidase inhibitory activity and anti-apoptotic properties for neurodegenerative diseases, such as PD, AD and ALS [2]. We incorporated the propargylamine moiety of rasagiline into the antioxidant-iron chelator moiety of an 8-hydroxyquinoline derivative of the iron chelating compound, VK28 [2, 3] to develop the multi target chelators M30 and HLA-20. N-propargyl functional group and its drug derivatives were shown in animal and cellular models of various neurodegenerative disorders with different insults that a series of propargyl derivatives exert significant neuroprotective and neurorescue activities [4-8]. The neuroprotection was ascribed mainly to a direct stabilization of the mitochondrial membrane potential and induction of anti-apoptotic pro-survival genes [8]. The novel multifunctional iron chelator, M30 was found to confer potential neuroprotective effects in preclinical neurodegenerative models with distinct etiologies, exerting selective iron chelation potency (compared with zinc and copper), radical scavenging, and inhibition of iron-induced membrane lipid peroxidation [2, 9]. M30 was shown to possess a significant neuroprotective, as well as neurorescue activities against the Parkinsonism-inducing neurotoxin N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity in mice [10, 11]. In addition, both M30 and VK28 were found to significantly improve behavioral performances and attenuate dopaminergic neuronal loss, proteasomal inhibition, iron accumulation, and microglial activation in the substantia nigra of mice injured with the proteasome inhibitor, lactacystin [12]. Furthermore, M30 treatment provided clear benefits in G93A-SOD-1 ALS mice, significantly increasing their survival and delaying the onset of neurological dysfunction [13]. In vitro studies in SH-SY5Y neuroblastoma, motor neuron-like NSC-34 and primary cortical cells demonstrated that M30 possesses multiple pharmacological activities, including improvement of neuronal survival in various neurotoxic models, induction of neuronal differentiation and up-regulation of hypoxia-inducible factor (HIF)-1 expression and HIF-1-target genes [13-17].

Neuroprotection by iron chelating agents has been widely attributed to their ability to prevent the iron from redox cycling and thereby, inhibit hydroxyl formation by the Fenton or Haber-Weiss reaction [1]. More recently, an additional level of neuroprotection by iron chelators has been postulated to involve inhibition of the activity of iron-dependent HIF-prolylhydroxylase (PHD) enzymes, resulting in the stabilization/activation of HIF-1 and the consequent activation of a broad set of HIF-1-target genes that may contribute to cell survival, iron regulation, and energy metabolism in the nervous system [18-22]. Indeed, it was demonstrated that desferoxamine (DFO) can activate HIF-1 and prevent neuronal death in both in vitro and in vivo models of ischemia, likely via inhibition of PHDs [23-25]. PHD inhibitors prevent oxidative cell death and ischemic injury, via activation of HIF-1-pathway [21]. Considering the diverse pharmacological properties of the novel iron chelator M30, the aim of our study was to identify distinct regulatory molecular mechanisms in the brain, that might be associated with the neuroprotective activity of the drug, including activation of HIF-1 signaling pathway and up-regulation of specific HIF-regulated target genes, expression of neurotrophic factors and antioxidant enzymes and induction of pro-survival cell signaling cascades.

MOLECULAR MECHANISM OF NEURORESTORATIVE ACTIVITY OF M30 AND MITOCHONDRIAL BIOGENESIS

Our previous studies have shown the novel multifunctional brain permeable iron, chelator M30 [5-(N-methyl-N-propargyaminomethyl)-8-hydroxyquinoline] and its piprezino derivative, HLA-20 possess neuroprotective, neurorescue and neurorestorative activities in vitro and in vivo, against several insults applicable to various neurodegenerative diseases, such as AD, PD, and ALS. We demonstrated that systemic chronic administration of M30 into mice resulted in up-regulation of hypoxia-inducible factor (HIF)-1 protein levels in various brain regions (e.g. cortex, striatum, and hippocampus) and spinal cord of adult mice. Real-time RT-PCR revealed that M30 differentially induced HIF-1-dependent target genes, including vascular endothelial growth factor (VEGF), erythropoietin (EPO), enolase-1, transferrin receptor (TfR), heme oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS), and glucose transporter (GLUT)-1. In addition, mRNA expression levels of the growth factors such as brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) and three antioxidant enzymes (catalase, superoxide dismutase SOD-1), and glutathione peroxidase (GPx) were up regulated by M30 treatment in a brain-region-dependent manner. Immunoblotting studies revealed that M30 induced a differentially enhanced phosphorylation of protein kinase C (PKC), mitogen-activated protein kinase (MAPK)/ERK kinase (MEK), protein kinase B (PKB/Akt), and glycogen synthase kinase-3 (GSK-3) (Fig. 1) [14].

Recently we have found 10 gene sets with previously unknown associations with the substantia nigra pars compacta of PD [26]. These gene sets pinpoint defects in mitochondrial electron transport, glucose utilization, and glucose sensing and reveal that they occur early in disease pathogenesis. Genes controlling cellular bioenergetics that are expressed in response to peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) are under expressed in patients with PD. Activation of PGC-1α results in increased expression of nuclear-encoded subunits of the mitochondrial respiratory chain and blocks the dopaminergic neuron loss induced by mutant α-synuclein or the pesticide rotenone in cellular disease models. Our systems biology analysis of PD has identified PGC-1α as a potential therapeutic target for early intervention since a defect in mitochondrial complex I has been shown in PD. Indeed we have recently shown that M30 and HLA-20, which possess neurodifferentiating, neurorescue and neurorestorative properties in vitro and in vivo [13, 22] activate PGC-1α, SIRT1(NAD-dependent protein deacetylase (neuroprotective), NFT (neurotrophic factor) and Tfam (mitochondrial transcription factor) in the hippocampal and cortical neurons in culture (Maoz et al, submitted for publication). These results indicate that the multi target M30 and HLA-20 initiate neuroprotection and neurorescue via biogenesis of mitochondria. Together, these results suggest that the multi target, iron chelator M30 can up-regulate a number of neuroprotective-adaptive mechanisms and pro-survival signaling pathways in the brain that might function as important disease modifying therapeutic targets for the drug in the context of neurodegenerative disease therapy (Fig. 2) [13-15].

The neuroprotective-neurorestorative mechanisms activated following M30 administration, are not completely understood. We have provided further insight into the various endogenous molecular mechanisms and prosurvival signaling pathways, activated in the brain following M30 systemic administration that might mediate neuroprotection. These include functional activation of HIF-1 signaling; regulation of a wide range of HIF-1-related protective genes, induction of mRNA expression levels of neurotrophic growth factors and antioxidant enzymes and upregulation of pro-survival signaling cascades. We have shown that the novel multifunctional compounds are strong chelators for iron and copper with higher selectivity for iron, and chelate iron (III) in a 3:1 M ratio, respectively [2, 27]. The fact that the new chelators have binding capability both for iron and copper, but with higher selectivity for iron may be important factors for the antioxidative-type drugs, since it is the excessive iron stores and iron-mediated generation of free radicals in the brain that are thought to be associated with neurodegenerative diseases [1, 2]. Therefore, the novel chelators with these properties would be expected to chelate iron instead of copper and hence would have potential use as drug candidates in neurodegenerative diseases. The current results demonstrate that M30 treatment produced a significant up-regulation of HIF-1 protein expression in the brain (e.g., cortex, striatum, and hippocampus and spinal cord). In addition, real time RT-PCR revealed that M30 differentially induced the transcription of a broad range of downstream HIF-1-related protective genes within the brain, such as those involved in erythropoiesis (EPO), angiogenesis (VEGF), glycolysis (Glut-1), and oxidative stress (HO-1), indicating a biological HIF-1 activation in the brain in response to M30 administration in vivo. This mechanism of HIF-1 up-regulation is consistent with previous studies demonstrating that iron chelators may function as hypoxia mimetic regulators; stabilizing and transactivating HIF-1, thus leading to the regulation of HIF-1-responsive genes [21, 24, 28, 29]. This may support adaptive mechanisms, which protect the brain from a hypoxic injury through regulation of cerebral metabolism and blood flow, promotion of angiogenesis, and induction of cytoprotection [20, 22, 30, 31]. Iron chelation by DFO enhanced HIF-1 activity and prevented neuronal death in both in vitro and in vivo models of ischemia via HIF-PHDs inhibition [12, 24, 29, 32, 33]. The protective effect of DFO against neuronal death after oxygen- and glucose-deprivation could be reversed by blockade of HIF-1 with antisense oligonucleotide transfection [12]. Thus, the activation of brain HIF-1 signal transduction pathway and consequent expression of HIF-1-target genes, possessing pro-survival properties, may implicate a link between M30-induced HIF-1-driven gene expression and neuroprotective capacities. Consequently, our in vitro findings demonstrated the ability of M30 to up-regulate HIF-1 and several HIF-1-target genes (e.g. enolase-1, VEGF, EPO, and p21) in cultured cortical neurons and NSC-34 cells, accompanied by protective effects against Aβ25-35- and mutant G93A-SOD-1-induced toxicity, respectively [13, 14, 34]. In vivo studies demonstrated that M30 significantly extended the survival of G93A-SOD-1 ALS mice and delayed the onset of the disease [13]. In addition, recent studies in APPswe/PSEN1 mouse model of AD have shown that M30 enhanced HIF-1 expression and reduced amyloid Aβ accumulation/plaque formation (mauscript in preparation). Activation of HIF-1 signaling pathway by M30 was also achieved in the peripheral organs (e.g., liver and heart). For example, of the HIF-1 target genes we examined in the liver, VEGF, enolase-1, TfR, iNOS, and GLUT-1 were significantly increased. Accordingly, activation of HIF-1 was recently shown to play a role in the effect of iron depletion by DFO on glucose metabolism in hepatocytes in vitro and in vivo [35]. In HepG2 cells, DFO stabilized HIF-1 and increased the expression of GLUT1 and insulin receptor. In addition, it was shown that DFO consistently increased the phosphorylation status of Akt/PKB and its targets FoxO1 and GSK-3, which mediate the effect of insulin on glucogenesis and glycogen synthesis, and up-regulated genes involved in glucose uptake and utilization. In vivo, iron depletion increased hepatic HIF-1 expression, GLUT-1 mRNA levels and Akt/PKB activity [35]. The specific activation of HIF-1 signaling and upregulation of HIF-1-related genes in the liver may be also associated with hepatic cytoprotection, as it was shown in various models of injury that stimulation of HIF system can protect the liver against apoptosis [36]. Another interesting finding in this study is the differential up-regulation of BDNF and GDNF in the CNS following M30 treatment. These data complement previous observations showing the ability of M30 and another multi-functional iron chelator drug, HLA20, to induce mRNA levels of BDNF in NSC-34 cells and cortical neurons [13, 34]. These drugs were also shown to promote neuronal differentiation, including cell body elongation, stimulation of neurite outgrowth and triggering cell cycle arrest in G0/G1 phase [16]. It was demonstrated that motor neuron differentiation, induced by M30 was modulated by the signaling inhibitors, PD98059 and GF109203X, indicating the involvement of MAPK and PKC pathways [13]. Additionally, in the current study we showed that M30 induced mRNA expression levels of the major antioxidant defense system, comprised of the antioxidant enzymes, catalase, SOD-1, and GPx, in various brain regions. These effects on transcriptional up-regulation of neuronal growth factors and antioxidant enzymes are presumably associated with the propargyl moiety, embedded in M30 molecule. Indeed, previous studies reported that several propargyl derivatives up-regulated mRNA expression of BDNF and NGF and increased protein levels of BDNF [37], suggesting that the stimulation of these neuronal survival pathways may provide an important step in their neuroprotective activity. In line with this, it was previously shown that propargylamines possess an antioxidant action and suppress the formation of free radicals by increasing the activity of the antioxidant enzymes, SOD and catalase in rat brain dopaminergic regions [22, 37]. By inducing antioxidant enzymes and decreasing the formation of reactive oxygen species, propargilamine-containing drugs may combat an oxidative challenge, implicated as a common causative factor in neurodegenerative diseases. Finally, M30 treatment induced a significant increase in brain expression of phosphorylated PKC, ERK1/2, AKT, and GSK-3. Regarding the role of these signal pathways in the regulation of neuroprotection, it has been reported in a number of studies that MEK/ERK and PI3K/AKT/GSK-3 pathways can promote cell survival, especially neuronal survival by both enhancing the expression of anti-apoptotic proteins and inhibiting the activity of pro-apoptotic ones [38-40]. In addition, these signaling pathways are well documented to play a key role in the regulation of HIF-1 [22] and thus, might be involved in the increased expression of HIF-1 following M30 treatment. Also, it cannot be ruled out that these signaling cascades are activated in the brain of M30-treated mice as a secondary phenomenon by an HIF-1-dependent gene product. Thus, considering the mechanism of action of M30, it can be assumed that the neuroprotective effects of the drug may be also associated with the activation of these pro-survival signaling cascades. Indeed, N-propargylamine and rasagiline confer neuroprotection and neurorescue effect via activation of PKC and MAPK pathways, coupled to pro-survival Bcl-2 family members and mitochondrial members stabilization [6, 37]. Although misregulation of the HIF pathway is only one component of a spectrum of reactions occurring in neurodegeneration, HIF-1 is a "master switch" being an important physiological response mechanism, likely resulting in several reproducible neuroprotective effects [41, 42]. Given the wide range and diversity of cellular functions regulated by the whole spectrum of HIF-1-target genes, it is suggested that this compensatory pathway mediated neuroprotection and is crucially involved in many physiological processes within the brain. Thus, in conclusion the novel therapeutic approach of pluri-potential multitatget iron chelating compounds, such as M30 and HLA-20 that targets a number of pharmacological sites involved in neurodegenerative processes and activates HIF pathway, mitochondrial biogenesis and downstream neuroprotective and neurotrophic genes, will broaden the current strategies for the treatment of neurological disorders such as PD and AD, and overall will open a new window for future development of drugs possessing a profound impact on neuron preservation, restoration and function.

  1. Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci 2004;5:863-873.
    Pubmed
  2. Zheng H, Gal S, Weiner LM, Bar-Am O, Warshawsky A, Fridkin M, Youdim MB. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant activity, prevention of lipid peroxide formation and monoamine oxidase inhibition. J Neurochem 2005;95:68-78.
    Pubmed
  3. Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB. Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology 2004;46:254-263.
    Pubmed
  4. Maruyama W, Akao Y, Carrillo MC, Kitani K, Youdium MB, Naoi M. Neuroprotection by propargylamines in Parkinson's disease: suppression of apoptosis and induction of prosurvival genes. Neurotoxicol Teratol 2002;24:675-682.
    Pubmed
  5. Bar-Am O, Yogev-Falach M, Amit T, Sagi Y, Youdim MB. Regulation of protein kinase C by the anti-Parkinson drug, MAO-B inhibitor, rasagiline and its derivatives, in vivo. J Neurochem 2004;89:1119-1125.
    Pubmed
  6. Weinreb O, Bar-Am O, Amit T, Chillag-Talmor O, Youdim MB. Neuroprotection via pro-survival protein kinase C isoforms associated with Bcl-2 family members. FASEB J 2004;18:1471-1473.
    Pubmed
  7. Youdim MB, Buccafusco JJ. Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders. Trends Pharmacol Sci 2005;26:27-35.
    Pubmed
  8. Naoi M, Maruyama W. Functional mechanism of neuroprotection by inhibitors of type B monoamine oxidase in Parkinson's disease. Expert Rev Neurother 2009;9:1233-1250.
    Pubmed
  9. Amit T, Avramovich-Tirosh Y, Youdim MB, Mandel S. Targeting multiple Alzheimer's disease etiologies with multimodal neuroprotective and neurorestorative iron chelators. FASEB J 2008;22:1296-1305.
    Pubmed
  10. Gal S, Zheng H, Fridkin M, Youdim MB. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. J Neurochem 2005;95:79-88.
    Pubmed
  11. Gal S, Zheng H, Fridkin M, Youdim MB. Restoration of nigrostriatal dopamine neurons in post-MPTP treatment by the novel multifunctional brain-permeable iron chelator-monoamine oxidase inhibitor drug, M30. Neurotox Res 2010;17:15-27.
    Pubmed
  12. Zhu W, Xie W, Pan T, Xu P, Fridkin M, Zheng H, Jankovic J, Youdim MB, Le W. Prevention and restoration of lactacystin-induced nigrostriatal dopamine neuron degeneration by novel brain-rmeable iron chelators. FASEB J 2007;21:3835-3844.
    Pubmed
  13. Kupershmidt L, Weinreb O, Amit T, Mandel S, Carri MT, Youdim MB. Neuroprotective and neuritogenic activities of novel multimodal iron-chelating drugs in motor-neuron-like NSC-34 cells and transgenic mouse model of amyotrophic lateral sclerosis. FASEB J 2009;23:3766-3779.
    Pubmed
  14. Kupershmidt L, Weinreb O, Amit T, Mandel S, Bar-Am O, Youdim MB. Novel molecular targets of the neuroprotective/neurorescue multimodal iron chelating drug M30 in the mouse brain. Neuroscience 2011;189:345-358.
    Pubmed
  15. Harten SK, Ashcroft M, Maxwell PH. Prolyl hydroxylase domain inhibitors: a route to HIF activationand neuroprotection. Antioxid Redox Signal 2010;12:459-480.
    Pubmed
  16. Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H, Fridkin M, Youdim MB. Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer's disease. J Neurochem 2007;100:490-502.
    Pubmed
  17. Avramovich-Tirosh Y, Reznichenko L, Mit T, Zheng H, Fridkin M, Weinreb O, Mandel S, Youdim MB. Neurorescue activity, APP regulation and amyloid-beta peptide reduction by novel multi-functionalbrain permeable iron- chelating- antioxidants, M-30 and green tea polyphenol, EGCG. Curr Alzheimer Res 2007;4:403-411.
    Pubmed
  18. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, Maxwell PH, Pugh CW, Ratcliffe PJ. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001;292:468-472.
    Pubmed
  19. Yu F, White SB, Zhao Q, Lee FS. HIF-1alpha binding to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc Natl Acad Sci U S A 2001;98:9630-9635.
    Pubmed
  20. Siddiq A, Aminova LR, Ratan RR. Hypoxia inducible factor prolyl 4-hydroxylase enzymes: center stage in the battle against hypoxia, metabolic compromise and oxidative stress. Neurochem Res 2007;32:931-946.
    Pubmed
  21. Siddiq A, Ayoub IA, Chavez JC, Aminova L, Shah S, LaManna JC, Patton SM, Connor JR, Cherny RA, Volitakis I, Bush AI, Langsetmo I, Seeley T, Gunzler V, Ratan RR. Hypoxia-inducible factor prolyl 4-hydroxylase inhibition. A target for neuroprotection in the central nervous system. J Biol Chem 2005;280:41732-41743.
    Pubmed
  22. Weinreb O, Amit T, Mandel S, Kupershmidt L, Youdim MB. Neuroprotective multifunctional iron chelators: from redox-sensitive process to novel therapeutic opportunities. Antioxid Redox Signal 2010;13:919-949.
    Pubmed
  23. Hurn PD, Koehler RC, Blizzard KK, Traystman RJ. Deferoxamine reduces early metabolic failure associated with severe cerebral ischemic acidosis in dogs. Stroke 1995;26:688-695.
    Pubmed
  24. Zaman K, Ryu H, Hall D, O'Donovan K, Lin KI, Miller MP, Marquis JC, Baraban JM, Semenza GL, Ratan RR. Protection from oxidative stress-induced apoptosis in cortical neuronal cultures by iron chelators is associated with enhanced DNA binding of hypoxia-inducible factor-1 and ATF-1/CREB and increased expression of glycolytic enzymes, p21(waf1/cip1), and erythropoietin. J Neurosci 1999;19:9821-9830.
    Pubmed
  25. Hamrick SE, McQuillen PS, Jiang X, Mu D, Madan A, Ferriero DM. A role for hypoxia-inducible factor-1alpha in desferoxamine neuroprotection. Neurosci Lett 2005;379:96-100.
    Pubmed
  26. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Grünblatt E, Moran LB, Mandel SA, Riederer P, Miller RM, Federoff HJ, Wüllner U, Papapetropoulos S, Youdim MB, Cantuti-Castelvetri I, Young AB, Vance JM, Davis RL, Hedreen JC, Adler CH, Beach TG, Graeber MB, Middleton FA, Rochet JC, Scherzer CR, Global PD, Gene Expression (GPEX) Consortium. PGC-1α, a potential therapeutic target for early intervention in Parkinson's disease. Sci Transl Med 2010;2:52ra73.
  27. Zheng H, Weiner LM, Bar-Am O, Epsztejn S, Cabantchik ZI, Warshawsky A, Youdim MB, Fridkin M. Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer's, Parkinson's, and other neurodegenerative diseases. Bioorg Med Chem 2005;13:773-783.
    Pubmed
  28. Wang GL, Semenza GL. Desferrioxamine induces erythropoietin gene expression and hypoxia-inducible factor 1 DNA-binding activity: implications for models of hypoxia signal transduction. Blood 1993;82:3610-3615.
    Pubmed
  29. Li YX, Ding SJ, Xiao L, Guo W, Zhan Q. Desferoxamine preconditioning protects against cerebral ischemia inrats by inducing expressions of hypoxia inducible factor 1 alpha and erythropoietin. Neurosci Bull 2008;24:89-95.
    Pubmed
  30. Sharp FR, Bernaudin M. HIF1 and oxygen sensing in the brain. Nat Rev Neurosci 2004;5:437-448.
    Pubmed
  31. Correia SC, Moreira PI. Hypoxia-inducible factor 1: a new hope to counteract neurodegeneration?. J Neurochem 2010;112:1-12.
    Pubmed
  32. Murphy TH, Schnaar RL, Coyle JT. Immature cortical neurons are uniquely sensitive to glutamate toxicity by inhibition of cystine uptake. FASEB J 1990;4:1624-1633.
    Pubmed
  33. Prass K, Ruscher K, Karsch M, Isaev N, Megow D, Priller J, Scharff A, Dirnagl U, Meisel A. Desferrioxamine induces delayed tolerance against cerebral ischemia in vivo and in vitro. J Cereb Blood Flow Metab 2002;22:520-525.
    Pubmed
  34. Avramovich-Tirosh Y, Bar-Am O, Amit T, Youdim MB, Weinreb O. Up-regulation of hypoxia-inducible factor (HIF)-1alpha and HIF-target genes in cortical neurons by the novel multifunctional iron chelator anti-Alzheimer drug, M30. Curr Alzheimer Res 2010;7:300-306.
    Pubmed
  35. Dongiovanni P, Valenti L, Ludovica Fracanzani A, Gatti S, Cairo G, Fargion S. Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol 2008;172:738-747.
    Pubmed
  36. Plock J, Frese S, Keogh A, Bisch-Knaden S, Ayuni E, Corazza N, Weikert C, Jakob S, Erni D, Dufour JF, Brunner T, Candinas D, Stroka D. Activation of non-ischemic, hypoxia-inducible signallingpathways up-regulate cytoprotective genes in the murine liver. J Hepatol 2007;47:538-545.
    Pubmed
  37. Weinreb O, Amit T, Bar-Am O, Youdim MB. Rasagiline:a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol 2010;92:330-344.
    Pubmed
  38. Koh SH, Kim SH, Kwon H, Park Y, Kim KS, Song CW, Kim J, Kim MH, Yu HJ, Henkel JS, Jung HK. Epigallocatechin gallate protects nerve growth factor differentiated PC12 cells from oxidative-radical-stress-induced apoptosis through its effect on phosphoinositide 3-kinase/Akt and glycogen synthase kinase-3. Brain Res Mol Brain Res 2003;118:72-81.
    Pubmed
  39. Risbud MV, Fertala J, Vresilovic EJ, Albert TJ, Shapiro IM. Nucleus pulposus cells upregulate PI3K/Akt and MEK/ERK signaling pathways under hypoxic conditions and resist apoptosis induced by serum withdrawal. Spine (Phila Pa 1976) 2005;30:882-889.
    Pubmed
  40. Sung SM, Jung DS, Kwon CH, Park JY, Kang SK, Kim YK. Hypoxia/reoxygenation stimulates proliferation through PKC-dependent activation of ERK and Akt in mouse neural progenitor cells. Neurochem Res 2007;32:1932-1939.
    Pubmed
  41. Bar Am O, Amit T, Youdim MB. Contrasting neuroprotective and neurotoxic actions of respective metabolites of anti-Parkinson drugs rasagiline and selegiline. Neurosci Lett 2004;355:169-172.
    Pubmed
  42. Bernhardt WM, Warnecke C, Willam C, Tanaka T, Wiesener MS, Eckardt KU. Organ protection by hypoxia and hypoxia-inducible factors. Methods Enzymol 2007;435:221-245.
    Pubmed