NADPH in Cell Death

0 downloads 0 Views 178KB Size Report
Roles of NAD+ / NADH and NADP+ / NADPH in Cell Death. Current Pharmaceutical ..... Alzheimer's and Parkinson's diseases and coenzyme Q10 as a po-.
12

Current Pharmaceutical Design, 2009, 15, 12-19

Roles of NAD+ / NADH and NADP+ / NADPH in Cell Death Weiliang Xia1,2, Zheng Wang4, Qing Wang6, Jin Han5, Cuiping Zhao1,2, Yunyi Hong5, Lili Zeng3, Le Tang3 and Weihai Ying1,2,* 1

Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; 2Med-X Research Institute, Shanghai Jiao Tong University; 3Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; 4Shanghai Institute of Digestive DiseasesRenji Hospital, Shanghai Jiao Tong University School of Medicine; 5School of Life Science and Biotechnology, Shanghai Jiao Tong University and 6Shanghai, P.R. China; Neurobiology Research Centre, Graduate school of Medicine, Faculty of Health & Behavioral Sciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia Abstract: A rapidly growing body of information has suggested that NAD (including NAD+ and NADH) and NADP (including NADP+ and NADPH) could be new fundamental factors in cell death: Many studies have indicated key roles of poly (ADP-ribose) polymerases and sirtuins --- two families of NAD-dependent enzymes --- in cell death; and NAD may also affect cell survival by influencing mitochondrial permeability transition, apoptosis-inducing factor and GAPDH. NAD may further influence cell survival by its effects on calcium homeostasis, gene expression and immunological functions. Due to the crucial roles of oxidative stress in cell death, NADPH may mediate cell death by its major effects on oxidative stress: NADPH is a key factor in cellular antioxidation systems; and NADPH oxidase is also a major generator of oxidative stress. With growing information about the novel biological properties of NAD and NADP, it is likely that new roles of NAD and NADP in cell death and various diseases will be elucidated. The elucidation may not only improve our understanding about the fundamental mechanisms of cell death, but also suggest new therapeutic targets for a variety of diseases.

Key Words: NAD, NADP, cell death, oxidative stress, mitochondria. 1. INTRODUCTION

2. ROLES OF NAD IN CELL DEATH

NAD, including nicotinamide adenine dinucleotide (NAD+) and reduced nicotinamide adenine dinucleotide (NADH), have been long known as key molecules in energy metabolism and mitochondrial functions. NADP, including nicotinamide adenine dinucleotide phosphate (NADP+) and reduced nicotinamide adenine dinucleotide phosphate (NADPH), have been known as critical molecules in antioxidation and reductive biosynthesis [1-3]. However, a rapidly growing body of information has suggested that NAD and NADP also play crucial roles in various biological processes, including calcium homeostasis, gene expression, immunological functions and aging [1-5].

2.1. Poly(ADP-Ribose) Polymerases (PARPs) in Cell Death

Due to its pivotal roles in a number of major diseases, cell death has been a research topic of intensive interest. With the increasing evidence suggesting that NAD and NADP could belong to the fundamental factors in cell death, it is warranted to generalize the information regarding the roles of NAD and NADP in cell death. Through this generalization, we may not only obtain improved understanding regarding NAD/NADP on cell death, but also identify future research directions on this topic.

*Address correspondence to this author at the Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200032, P.R. China; Tel: 86-21-62932302; Fax: 86-21-62932302; E-mail: [email protected]

1381-6128/09 $55.00+.00

PARPs are a family of enzymes that consume NAD+ to produce poly(ADP-ribose) (PAR) on target proteins [6]. PARP-1 has been the most intensively studied member of PARP family, which plays important roles in a variety of biological processes, including DNA repair, gene expression, genomic stability, cell cycle, and cell death [4, 6, 7]. A number of studies have demonstrated that excessive PARP-1 activation mediates cell death induced by oxidative stress under many conditions [8, 9]. In animal model studies, PARP-1 has been indicated to play critical roles in many diseases, including ischemic brain damage [10, 11], Parkinson’s disease (PD) [12-14] and diabetes [15-19]. For examples, both pharmacological and genetic inhibition of PARP-1 can profoundly decrease infarct formation in animal models of brain ischemia [9, 20]; PARP-1 activation has also been shown to mediate the neuronal death induced by MPTP, a model toxin for PD, both in vitro [21, 22] and in vivo [2325]; and PARP-1 activation mediates -amyloid-induced neuronal death, which is an in vitro model of Alzheimer’s disease (AD) [26, 27]. A number of studies have further suggested that PARP-1 activation plays important roles in traumatic brain injury [28], diabetes [29], hypoglycemic brain injury [30], and shock and inflammation [6, 31]. Due to the-

© 2009 Bentham Science Publishers Ltd.

Roles of NAD + / NADH and NADP+ / NADPH in Cell Death

se lines of findings, PARP-1 has become a therapeutic target for various diseases [6, 31, 32]. We have provided direct evidence indicating that NAD+ depletion mediates PARP-1-induced cell death [33, 34]. It has also been indicated that MPT [34] and apoptosisinducing factor (AIF) translocation [35] link NAD+ depletion to cell death. New mechanisms regarding PARP-1 cytotoxicity have also been suggested by several lines of studies: First, ADP-ribose, that can be generated by PARP-1/PARG, is capable of causing TRPM2 opening, leading to elevated intracellular calcium concentrations and cell death [27, 36, 37]; however, it has also been suggested that PAR, instead ADP-ribose monomers, mediates PARP-1-induced AIF translocation and cell death [38, 39]. Second, it was suggested that NAD+ depletion causes cell death by decreasing SIRT1 activity [40]. Third, c-Jun N-terminal kinases (JNKs) could contribute to PARP-1-mediated cell death [41-44]; and extracellular signal-regulated kinases 1/2 (ERK1/2) can induce PARP-1 activation by directly phosphorylating the enzyme [41]. Cumulative evidence has suggested that other PARPs could also play significant roles in cell injury: It was reported that PARP-2 activity is detrimental in an animal model of focal brain ischemia, while it is beneficial in a model of global ischemia [45]; a recent study has suggested that PARP-2 mediates the survival of CD4+CD8+ doublepositive T cells during thymopoiesis [46]; and it has also been found that overexpression of tankyrase 2 can cause rapid cell death [47]. Future studies are warranted to further determine the interactions among PARPs under various physiological and pathological conditions. 2.2. Sirtuins in Cell Death It has been demonstrated that Sir2 is a key enzyme mediating the life span of yeast and C. elegans [48]: A decrease or increase in gene copy of Sir2 shortens or extends the replicative life span of yeast, respectively [49]; and increased gene copy of the Sir2 gene homolog in C. elegans also extends its life span [50]. Sir2 is a NAD+-dependent histone deacetylase, which produces protein deacetylation by consuming NAD+. Sirtuins are the mammalian homologs of Sir2, which include seven members, i.e., SIRT1-7. A number of studies have indicated that sirtuins are novel, key factors in cell death under many conditions. SIRT1 is the major subject in a majority of published studies regarding the roles of sirtuins in cell death. Most of these studies have suggested that SIRT1 activity is beneficial for cell survival. For example, it has been suggested the nicotinamide mononucleotide adenylyltransferases-1 (NMNAT-1) in the Wallerian degeneration slow (Wld(S)) protein could mediate the protective effects of the Wallerian mutation against axonal degeneration [51, 52]; both SIRT1 overexpression and administration of the SIRT1 activator resveratrol can decrease -amyloid-induced NF-B signaling and produce neuroprotective effects [53]; and SIRT1 deficiency can lead to significant increases in PARP-1 activity, causing AIF-mediated cell death [54]. Several mechanisms regarding the roles of SIRT1 in cell death have been suggested: First, SIRT1 may enhance cell survival by decreasing acetylation of p53 thus enhancing p53 degradation [55]; sec-

Current Pharmaceutical Design, 2009, Vol. 15, No. 1

13

ond, SIRT1 may also reduce cell death by preventing PARP1 activation [54]; and third, SIRT1 may indirectly reduce microglia-induced neurotoxicity by producing deacetylation of NFB [53]. However, SIRT1 could also be detrimental to cell survival under certain conditions: SIRT1 can deacetylate NFB thus enhancing the susceptibility of cells to TNF toxicity [56]. SIRT2 is a tubulin deacetylase. It has been reported that SIRT2 inhibitors rescues alpha-synuclein-mediated neurotoxicity in a PD model [57]. Activation of SIRT2 by resveratrol was also shown to abolish the protective effects of Wld(s) protein against axonal degeneration [58]. Thus, in contrast to the mainly beneficial roles of SIRT1 activity in cell survival, current studies have suggested that SIRT2 produces deleterious effects on cell survival under some conditions. SIRT3 is a mitochondrial deacetylase [59]. It has been reported that SIRT3 can reduce stress-induced apoptosis of cardiomyocytes by the following pathway: SIRT3 deacetylates Ku70, which promotes the binding of Ku70 to Bax thus preventing Bax translocation into the mitochondria [60]. It has also been reported that fasting-induced increases in NMNAT expression require mitochondrial presence of SIRT3 and SIRT4 to decrease stress-induced cell apoptosis [61]. However, in contrast to these observations suggesting beneficial roles of SIRT3 activity in cell survival, it has been reported that SIRT3 is pro-apoptotic and contributes to the JNK2-mediated apoptosis [62]. In summary, sirtuins have rapidly emerged as critical factors in cell survival. These enzymes appear to be the key links among NAD, p53, NFB, aging and cell death. While SIRT1 appears to be largely beneficial to cell survival, SIRT2 seems to be mainly detrimental to cell survival. Just like such factors as nitric oxide and NFB, sirtuins can be either beneficial or detrimental to cell survival. It is expected that future studies on the roles of sirtuins in cell death would provide ample information that would profoundly deepen our understanding regarding the mechanisms of cell death. 2.3. NAD in Apoptosis There have been only small number of studies regarding the roles of NAD in apoptosis, which have suggested that NAD may be involved in apoptosis: NADH / NADPH depletion is an early event in apoptosis [63]; and selective inhibitors of NAD+ synthesis can induce apoptosis [64]. There are several potential mechanisms underlying the roles of NAD in apoptosis: 1) NADH / NAD+ ratio is a major index of cellular reducing power that affects MPT, which mediates apoptosis under many conditions [65]; 2) NAD plays a key role in energy metabolism that is a key factor determining cell death modes; 3) NAD+-dependent sirtuins may mediate apoptosis [56]; and 4) NAD+ levels significantly affect the activity of caspase-dependent endonuclease DFF40 --- an executioner of DNA fragmentation in certain apoptotic cascades [66]. 2.4. NAD, Oxidative Stress and Cell Death Due to the critical roles of oxidative stress in cell death, NAD may affect cell survival by influencing oxidative stress and antioxidation systems in cells: First, NAD+ can be converted by NAD kinases to NADP+ --- the precursor for

14 Current Pharmaceutical Design, 2009, Vol. 15, No. 1

NADPH formation [67]; second, NADH / NAD + ratio is an index of cellular reducing potential, since the redox couple plays key roles in numerous redox reactions; third, some studies have suggested that NADH can produce direct antioxidation effects [68-70]; and fourth, NAD+ can inhibit ROS generation from -ketoglutarate dehydrogenase and pyruvate dehydrogenase [71]. It is noteworthy that excessive intracellular NADH can produce ‘reductive stress’, which may result from the capacity of xanthine oxidase / xanthine dehydrogenase to generate ROS by oxidizing NADH [72], or from the capacity of NADH to induce release of ferrous iron from ferritin [66]. 2.5. AIF and GAPDH in Cell Death AIF is a NADH oxidase, which can act as both a prodeath factor and a pro-survival factor [73-75]. Nuclear translocation of AIF from mitochondria is a key step in both caspase-independent apoptosis [74] and PARP-1 cytotoxicity [35]. However, AIF also plays an important role in mitochondrial Complex I activity [76]; and genetic deletion of AIF can produce dilated cardiomyopathy, skeleton muscle atrophy, and neurodegeneration [74]. Because it has been indicated that AIF is a NADH oxidase and prevention of NAD+ depletion can block PARP-1-mediated nuclear translocation of AIF [34], it is highly likely that NAD mediates the activity of AIF. Our latest study has found that aurintricarboxylic acid --- a Ca2+-Mg2+-endonuclease inhibitor --can abolish DNA alkylating agent-induced nuclear condensation in astrocytes, despite nuclear translocation of AIF (unpublished finding). This observation suggests that at least under certain conditions, it may be insufficient for nuclear AIF translocation itself to induce nuclear condensation. NAD is the major co-factor of GAPDH. GAPDH has been established as a mediator of apoptosis under many conditions. It has been found that GAPDH binds Siah which is then translocated into nucleus to mediate apoptosis [77-80]. 2.6. NAD in Axonal Degeneration Axonal degeneration is a key pathological change in multiple neurological diseases [81]. Wallerian degeneration slow (Wld(S)) mouse model is a widely used model for studying the mechanisms of axonal degeneration [81]. The mutation of Wld(S) mice leads to overexpression of a chimeric protein --- Wld(S) protein that consists of the key NAD+-synthesizing enzyme NMNAT-1 and the ubiquitin assembly protein Ufd2a. The overexpression of Wld(S) protein can cause delay of injury-induced axonal degeneration. It has been suggested that the NMNAT-1 in the Wld(S) protein could mediate the protective effects of the Wallerian mutation by affecting SIRT1 --- a member of sirtuins [51], or by preventing NAD+ loss in degenerating axons [52]. However, there are also studies suggesting that NMNAT-1 itself may be insufficient to produce the protective effects of the Wld(S) protein [82, 83]. 3 ROLES OF NADP IN CELL DEATH Oxidative stress is a key factor in cell death. Because NADP metabolism plays critical roles in both antioxidation system and generation of oxidative stress, NADP could play important roles in cell death.

Ying et al.

NADPH is a key factor in cellular antioxidation through the following mechanisms: First, GSH is essential for the functions of such key antioxidation enzymes as glutathione peroxidase [84], while NADPH is required for regeneration of GSH from GSSG; second, NADPH is a key component in another important antioxidation system --- thioredoxin system [85]; and third, in such cell types as red cells NADPH binds the important H2O2-disposing enzyme catalase [86], which can reactivate catalase when catalase is inactivated by H2O2. Cumulating evidence has demonstrated that NADPH can also significantly contribute to generation of oxidative stress through the activity of NADPH oxidase that generates superoxide from oxygen and NADPH. NADPH oxidase activity is present in not only phagocytes, but also various tissues and cell types [87, 88]. There are seven members of NOX family of NADPH oxidase, including phagocyte NADPH oxidase itself (NOX2/gp91(phox)) and six homologs of the cytochrome subunit of the phagocyte NADPH oxidase, i.e., NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2 [87]. These enzymes share the capacity to transport electrons across the plasma membrane, resulting in superoxide generation, which can be regulated by such factors as small guanosine triphosphatase Rac, protein kinase C and Ca2+ [87-90]. Many studies have indicated NADPH oxidase as a key factor in cell death under both in vitro and in vivo conditions [91]. For examples, NADPH oxidase appears to play a key role in the ROS generation in the neurons exposed to oxygen-glucose deprivation --- an in vitro model for brain ischemia [92]; both genetic and pharmacological inhibition of NADPH oxidase is protective against ischemic brain injury [93]; and the NADPH oxidase activation in astrocytes was shown to mediate -amyloid-induced neuronal death [94]. Interestingly, it has been reported that ischemia can induce NOX2 expression at the nucleus of cardiomyocytes, which appears to mediate ischemia-induced apoptosis [95]. Because numerous studies have indicated critical roles of oxidative stress in cell death [96], it is expected that many future studies would indicate important roles of NADPH oxidase and other NADPH-related enzymes in various diseases. 4. THERAPEUTIC POTENTIAL OF NAD AND NADP 4.1. Therapeutic Potential of NAD + Our cell culture studies have provided the first evidence indicating that NAD+ treatment can abolish PARP-1-induced astrocyte death [33, 34, 97, 98]. Recent in vitro studies have also shown that NAD + treatment can attenuate OGD-induced neuronal death [99] and PARP-1-mediated myocyte death [40]. These results suggest that NAD+ may be used in vivo to decrease PARP-1-mediated tissue injury. We have used a rat model of transient focal ischemia to test our hypothesis that NAD+ administration can decrease ischemic brain injury [100]: Intranasal delivery with NAD+ at 2 hrs after ischemic onset was shown to reduce infarct formation by nearly 90%, and to significantly decrease the ischemia/reperfusioninduced neurological deficits. These observations have provided the first in vivo evidence that that NAD+ administration may be a new strategy for treating stroke patients, and

Roles of NAD + / NADH and NADP+ / NADPH in Cell Death

NAD+ metabolism is a novel target for decreasing ischemic brain damage. NAD+ may have its unique merits as a cytoprotective agent: 1) NAD+ may reduce cell death not only by blocking PARP-1 toxicity, but also by enhancing energy metabolism and SIRT-1 activity; 2) NAD + is protective even when applied at 3-4 hrs after PARP-1 activation, suggesting that NAD+ administration may have long window of opportunity in decreasing tissue injury; and 3) compared with other cytoprotective factors as pyruvate, NAD+ can produce greatest protective effects against PARP-1 cytotoxicity in vitro [33, 34]. It is noteworthy that NAD+ administration may decrease the brain injury in not only cerebral ischemia, but also many other diseases [101] due to the following reasons: NAD + treatment can profoundly decrease PARP-1-mediated cell injury; and pathological roles of PARP-1 have been implicated in many diseases such as diabetes, PD and AD [5]. Several studies have reported that NADH administration can produce beneficial effects in treating PD [102, 103], which may be accounted for partially by the capacity of NADH to increase bioavailability of plasma levodopa. NADH administration may also be used for treating AD patients, since NADH can improve cognitive functions [104]. Our studies have provided direct evidence showing that NADH treatment can decrease the cell death induced by the DNA alkylating agent MNNG [98], thus providing certain explanations for the beneficial effects of NADH. However, our cell culture studies have also suggested that NADH treatment can be cytotoxic when cells are exposed to the peroxynitrite generator SIN-1 (data not shown), in contrast to the beneficial effects of NAD+ administration against SIN1 toxicity. These observations not only highlight the merits of NAD+ as a cytoprotective agent, but also suggest that great caution should be taken when applying NADH for treating diseases. 4.2. Therapeutic Potential of NAD + Precursors Nicotinamide administration can decrease tissue injury in the animal models of cerebral ischemia [105-108], spinal cord injury [109], PD [110] and multiple sclerosis [111]. Cell culture studies have also shown the cytoprotective effects of nicotinamide against oxidative stress and oxygen-

Fig. (1). The pathways from NAD isoforms to cell death are shown.

Current Pharmaceutical Design, 2009, Vol. 15, No. 1

15

glucose deprivation [112, 113]. The protective effects may result from the capacity of nicotinamide to restore NAD+ levels [105, 114], inhibit PARP-1 [6, 114], activate Akt1 [115] and block mitochondrial permeability transition [113, 116]. However, nicotinamide may also impair cell survival and longevity, due to its inhibitory effects on sirtuins [117]. Recent studies have also shown that nicotinamide riboside -- a new NAD+ precursor in eukaryotes --- can enhance Sir2dependent gene silencing and extend the replicative lifespan of yeast without calorie restriction [118]. The beneficial effects of nicotinamide riboside appear to result from the capacity of nicotinamide riboside to increase NAD + synthesis [118]. Increasing evidence has also suggested significant biological activities of nicotinic acid --- an important component in NAD+ metabolism: Nicotinic acid can significantly affect brain functions by such pathways as inducing glutamate release [119]. 4.3. Therapeutic Potential of NADPH Oxidase It appears to be increasingly important for cell survival to maintain NADPH as a ‘good guy’, since it can act as either a ‘good guy’ or a ‘bad guy’ in cellular antioxidation systems. It is of particular importance to modulate NADPH-related properties to reduce the detrimental effects of NADPH in diseases, because NADPH oxidase can play critical pathological roles in multiple illnesses. Several strategies may be used to modulate NADPH oxidase activity: 1) Directly inhibit NADPH oxidase by using NADPH oxidase inhibitors; 2) inhibit excessive NADPH generation by modulating the multiple NADPH-generating reactions; 3) maintain the activities of such NADPH-consuming enzymes as glutathione reductase, so as to ensure efficient NADPH flux through these pathways, resulting in prevention of excessive NADPH supply to NADPH oxidase; and 4) indirectly inhibit NADPH oxidase by manipulating such intracellular modulators of the enzyme as the small guanosine triphosphatase Rac [87, 88, 90]. 5. SUMMARY Based on the above discussion, it is increasingly clear that NAD and NADP play critical roles in cell death, which can affect cell survival through several pathways (Fig. 1). It

16 Current Pharmaceutical Design, 2009, Vol. 15, No. 1

is expected that a growing number of pathways would be elucidated by future studies, which would further highlight the central roles of NAD and NADP in linking cell survival with various biological properties, including energy metabolism, mitochondrial functions, calcium homeostasais, oxidative stress/antioxidation and gene expression. Due to the pivotal roles of aberrant cell death in multiple major illnesses, including stroke, myocardial ischemia, cancer, Alzheimer’s disease, and diabetes, it is conceivable that NADand NADP-dependent pathways would become increasingly attractive therapeutic targets for the diseases.

Ying et al.

[10] [11] [12] [13] [14]

[15]

ACKNOWLEDGMENTS This work is supported by a Key Grant for Basic Research of Shanghai Municipal Scientific Committee (Grant# 08JC1415400, to W.Ying), by a Key Grant of Shanghai Municipal Educational Committee (to W.Ying), by the Shanghai Med-X Engineering & Technology Research Center Grant for Physical Therapy and Diagnostics of Major Diseases (Grant# 08DZ2211200), by the ‘Start-Up Funds from 985 Project’ (to W.Ying), and by Shanghai Municipal Scientific Committee (06JC14052).

[16]

[17]

[18]

[19]

ABBREVIATIONS AD

= Alzheimer’s disease

AIF

= Apoptosis-inducing factor

ERK

= Extracellular signal-regulated kinase

JNK

= c-Jun N-terminal kinase

NAD

= Nicotinamide adenine dinucleotide

NADP

= Nicotinamide adenine dinucleotide phosphate

PARP

= Poly(ADP-ribose) polymerases

PD

= Parkinson’s disease

NMNAT-1 = Nicotinamide mononucleotide adenylyltransferases-1

[20]

[21]

[22]

[23]

[24]

REFERENCES [1]

[2] [3]

[4] [5] [6] [7]

[8]

[9]

Berger F, Ramirez-Hernandez MH, Ziegler M. The new life of a centenarian: signalling functions of NAD(P). Trends Biochem Sci 2004; 29: 111-8. Belenky P, Bogan KL, Brenner C. NAD+ metabolism in health and disease. Trends Biochem Sci 2007; 32: 12-9. Pollak N, Dolle C, Ziegler M. The power to reduce: pyridine nucleotides--small molecules with a multitude of functions. Biochem J 2007; 402: 205-18. Ying W. NAD+ and NADH in cellular functions and cell death. Front Biosci 2006; 11: 3129-48. Ying W. NAD+ and NADH in brain functions, brain diseases and brain aging. Front in Biosci 2007; 12: 1863-88. Virag L, Szabo C. The therapeutic potential of poly(ADP-ribose) polymerase inhibitors. Pharmacol Rev 2002; 54: 375-429. Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 2006; 7: 517-28. Zhang J, Dawson VL, Dawson TM, Snyder SH. Nitric oxide activation of poly(ADP-ribose) synthetase in neurotoxicity. Science 1994; 263: 687-9. Eliasson MJ, Sampei K, Mandir AS, Hurn PD, Traystman RJ,Bao J, Pieper A, Wang ZQ, Dawson TM, Snyder SH, Dawson VL.

[25]

[26]

[27]

[28]

[29]

Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med 1997; 3: 1089-95. Chan PH. Role of oxidants in ischemic brain damage. Stroke 1996; 27: 1124-9. Chan PH. Reactive oxygen radicals in signaling and damage in the ischemic brain. J Cereb Blood Flow Metab 2001; 21: 2-14. Ying W. Deleterious network hypothesis of aging. Med Hypotheses 1997; 48: 143-8. Wolozin B, Golts N. Iron and Parkinson's disease. Neuroscientist 2002; 8: 22-32. Beal MF. Mitochondrial dysfunction and oxidative damage in Alzheimer's and Parkinson's diseases and coenzyme Q10 as a potential treatment. J Bioenerg Biomembr 2004; 36: 381-6. Ying W. A new hypothesis of neurodegenerative diseases: the deleterious network hypothesis. Med Hypotheses 1996; 47: 307-13. Moreira PI, Honda K, Liu Q, Santos MS, Oliveira CR, Aliev G, et al. Oxidative stress: the old enemy in Alzheimer's disease pathophysiology. Curr Alzheimer Res 2005; 2: 403-8. Zhu X, Raina AK, Lee HG, Casadesus G, Smith MA, Perry G. Oxidative stress signalling in Alzheimer's disease. Brain Res 2004; 1000: 32-9. Mhatre M, Floyd RA, Hensley K. Oxidative stress and neuroinflammation in Alzheimer's disease and amyotrophic lateral sclerosis: common links and potential therapeutic targets. J Alzheimers Dis 2004; 6: 147-57. Keller JN, Guo Q, Holtsberg FW, Bruce-Keller AJ, Mattson MP. Increased sensitivity to mitochondrial toxin-induced apoptosis in neural cells expressing mutant presenilin-1 is linked to perturbed calcium homeostasis and enhanced oxyradical production. J Neurosci 1998; 18: 4439-50. Endres M, Wang ZQ, Namura S, Waeber C, Moskowitz MA. Ischemic brain injury is mediated by the activation of poly(ADPribose)polymerase. J Cereb Blood Flow Metab 1997; 17: 1143-51. Cosi C, Colpaert F, Koek W, Degryse A, Marien M. Poly(ADPribose) polymerase inhibitors protect against MPTP-induced depletions of striatal dopamine and cortical noradrenaline in C57B1/6 mice. Brain Res 1996; 729: 264-9. Mandir AS, Simbulan-Rosenthal CM, Poitras MF, Lumpkin JR, Dawson VL, Smulson ME, et al. A novel in vivo post-translational modification of p53 by PARP-1 in MPTP-induced parkinsonism. J Neurochem 2002; 83: 186-92. Mandir AS, Przedborski S, Jackson-Lewis V, Wang ZQ, SimbulanRosenthal CM, Smulson ME, et al. Poly(ADP-ribose) polymerase activation mediates 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced parkinsonism. Proc Natl Acad Sci USA 1999; 96: 5774-9. Iwashita A, Yamazaki S, Mihara K, Hattori K, Yamamoto H, Ishida J, et al. Neuroprotective effects of a novel poly(ADP-ribose) polymerase-1 inhibitor, 2-[3-[4-(4-chlorophenyl)-1-piperazinyl] propyl]-4(3H)-quinazolinone (FR255595), in an in vitro model of cell death and in mouse 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine model of Parkinson's disease. J Pharmacol Exp Ther 2004; 309: 1067-78. Przedborski S, Jackson-Lewis V, Djaldetti R, Liberatore G, Vila M, Vukosavic S, et al. The parkinsonian toxin MPTP: action and mechanism. Restor Neurol Neurosci 2000; 16: 135-42. Hensley K, Butterfield DA, Hall N, Cole P, Subramaniam R, Mark R, et al. Reactive oxygen species as causal agents in the neurotoxicity of the Alzheimer's disease-associated amyloid beta peptide. Ann N Y Acad Sci 1996; 786: 120-34. Fonfria E, Marshall IC, Boyfield I, Skaper SD, Hughes JP, Owen DE, et al. Amyloid beta-peptide(1-42) and hydrogen peroxideinduced toxicity are mediated by TRPM2 in rat primary striatal cultures. J Neurochem 2005; 95: 715-23. LaPlaca MC, Zhang J, Raghupathi R, Li JH, Smith F, Bareyre FM, et al. Pharmacologic inhibition of poly(ADP-ribose) polymerase is neuroprotective following traumatic brain injury in rats. J Neurotrauma 2001; 18: 369-76. Pieper AA, Brat DJ, Krug DK, Watkins CC, Gupta A, Blackshaw S, et al. Poly(ADP-ribose) polymerase-deficient mice are protected from streptozotocin-induced diabetes. Proc Natl Acad Sci USA 1999; 96: 3059-64.

Roles of NAD + / NADH and NADP+ / NADPH in Cell Death [30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48] [49]

Suh SW, Aoyama K, Chen Y, Garnier P, Matsumori Y, Gum E, et al. Hypoglycemic neuronal death and cognitive impairment are prevented by poly(ADP-ribose) polymerase inhibitors administered after hypoglycemia. J Neurosci 2003; 23: 10681-90. Szabo C, Dawson VL. Role of poly(ADP-ribose) synthetase in inflammation and ischaemia- reperfusion. Trends Pharmacol Sci 1998; 19: 287-98. Pieper AA, Verma A, Zhang J, Snyder SH. Poly (ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol Sci 1999; 20: 171-81. Ying W, Garnier P, Swanson RA. NAD+ repletion prevents PARP1-induced glycolytic blockade and cell death in cultured mouse astrocytes. Biochem Biophys Res Commun 2003; 308: 809-13. Alano CC, Ying W, Swanson RA. Poly(ADP-ribose) polymerase1-mediated cell death in astrocytes requires NAD+ depletion and mitochondrial permeability transition. J Biol Chem 2004; 279: 18895-902. Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002; 297: 259-63. Fonfria E, Marshall IC, Benham CD, Boyfield I, Brown JD, Hill K, et al. TRPM2 channel opening in response to oxidative stress is dependent on activation of poly(ADP-ribose) polymerase. Br J Pharmacol 2004; 143: 186-92. Yang KT, Chang WL, Yang PC, Chien CL, Lai MS, Su MJ, et al. Activation of the transient receptor potential M2 channel and poly(ADP-ribose) polymerase is involved in oxidative stressinduced cardiomyocyte death. Cell Death Differ2005; 13:1815-26. Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, et al. Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci USA 2006; 103: 18308-13. Yu SW, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM, et al. Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci USA 2006; 103: 18314-9. Pillai JB, Isbatan A, Imai S, Gupta MP. Poly(ADP-ribose) polymerase-1-dependent cardiac myocyte cell death during heart failure is mediated by NAD+ depletion and reduced Sir2alpha deacetylase activity. J Biol Chem 2005; 280: 43121-30. Kauppinen TM, Chan WY, Suh SW, Wiggins AK, Huang EJ, et al. Direct phosphorylation and regulation of poly(ADP-ribose) polymerase-1 by extracellular signal-regulated kinases 1/2. Proc Natl Acad Sci USA 2006; 103: 7136-41. Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, et al. DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell 2007; 25: 297-308. Xu Y, Huang S, Liu ZG, Han J. Poly(ADP-ribose) polymerase-1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2-mediated JNK1 activation. J Biol Chem 2006; 281: 8788-95. Zhang S, Lin Y, Kim YS, Hande MP, Liu ZG, Shen HM. c-Jun Nterminal kinase mediates hydrogen peroxide-induced cell death via sustained poly(ADP-ribose) polymerase-1 activation. Cell Death Differ 2007; 14: 1001-10. Kofler J, Otsuka T, Zhang Z, Noppens R, Grafe MR, Koh DW, et al. Differential effect of PARP-2 deletion on brain injury after focal and global cerebral ischemia. J Cereb Blood Flow Metab 2006; 26: 135-41. Yelamos J, Monreal Y, Saenz L, Aguado E, Schreiber V, Mota R, et al. PARP-2 deficiency affects the survival of CD4+CD8+ doublepositive thymocytes. EMBO J 2006; 25: 4350-60. Kaminker PG, Kim SH, Taylor RD, Zebarjadian Y, Funk WD, Morin GB, et al. TANK2, a new TRF1-associated poly(ADPribose) polymerase, causes rapid induction of cell death upon overexpression. J Biol Chem 2001; 276: 35891-9. Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem 2004; 73: 417-35. Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev 1999; 13: 2570-80.

Current Pharmaceutical Design, 2009, Vol. 15, No. 1 [50]

[51]

[52]

[53]

[54]

[55]

[56]

[57]

[58]

[59]

[60]

[61]

[62] [63]

[64]

[65] [66]

[67] [68] [69]

[70]

[71]

17

Tissenbaum HA, Guarente L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 2001; 410: 22730. Araki T, Sasaki Y, Milbrandt J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science 2004; 305: 1010-3. Wang J, Zhai Q, Chen Y, Lin E, Gu W, McBurney MW, et al. A local mechanism mediates NAD-dependent protection of axon degeneration. J Cell Biol 2005; 170: 349-55. Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi S, et al. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem 2005; 280: 40364-74. Kolthur-Seetharam U, Dantzer F, McBurney MW, de Murcia G, Sassone-Corsi P. Control of AIF-mediated Cell Death by the Functional Interplay of SIRT1 and PARP-1 in Response to DNA Damage. Cell Cycle 2006; 5: 873-7. Kume S, Haneda M, Kanasaki K, Sugimoto T, Araki S, Isono M, et al. Silent information regulator 2 (SIRT1) attenuates oxidative stress-induced mesangial cell apoptosis via p53 deacetylation. Free Radic Biol Med 2006; 40: 2175-82. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 2004; 23: 2369-80. Outeiro TF, Kontopoulos E, Altmann SM, Kufareva I, Strathearn KE, Amore AM, et al. Sirtuin 2 inhibitors rescue alpha-synucleinmediated toxicity in models of Parkinson's disease. Science 2007; 317: 516-9. Suzuki K, Koike T. Mammalian Sir2-related protein (SIRT) 2mediated modulation of resistance to axonal degeneration in slow Wallerian degeneration mice: a crucial role of tubulin deacetylation. Neuroscience 2007; 147: 599-612. Hallows WC, Albaugh BN, Denu JM. Where in the cell is SIRT3?-functional localization of an NAD+-dependent protein deacetylase. Biochem J 2008; 411: e11-3. Sundaresan NR, Samant SA, Pillai VB, Rajamohan SB, Gupta MP. SIRT3 is a stress-responsive deacetylase in cardiomyocytes that protects cells from stress-mediated cell death by deacetylation of Ku70. Mol Cell Biol 2008; 28: 6384-401. Yang H, Yang T, Baur JA, Perez E, Matsui T, Carmona JJ, et al. Nutrient-sensitive mitochondrial NAD+ levels dictate cell survival. Cell 2007; 130: 1095-107. Allison SJ, Milner J. SIRT3 is pro-apoptotic and participates in distinct basal apoptotic pathways. Cell Cycle 2007; 6: 2669-77. Gendron MC, Schrantz N, Metivier D, Kroemer G, Maciorowska Z, Sureau F, et al. Oxidation of pyridine nucleotides during Fasand ceramide-induced apoptosis in Jurkat cells: correlation with changes in mitochondria, glutathione depletion, intracellular acidification and caspase 3 activation. Biochem J 2001; 353: 357-67. Hasmann M, Schemainda I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res 2003; 63: 7436-42. Zoratti M, Szabo I. The mitochondrial permeability transition. Biochim Biophys Acta 1995; 1241: 139-76. Jaeschke H, Kleinwaechter C, Wendel A. NADH-dependent reductive stress and ferritin-bound iron in allyl alcohol-induced lipid peroxidation in vivo: the protective effect of vitamin E. Chem Biol Interact 1992; 81: 57-68. McGuinness ET, Butler JR. NAD+ kinase--a review. Int J Biochem 1985; 17: 1-11. Kirsch M, De Groot H. NAD(P)H, a directly operating antioxidant? FASEB J 2001; 15: 1569-74. Mazzio EA, Soliman KF. Cytoprotection of pyruvic acid and reduced beta-nicotinamide adenine dinucleotide against hydrogen peroxide toxicity in neuroblastoma cells. Neurochem Res 2003; 28: 733-41. Olek RA, Ziolkowski W, Kaczor JJ, Greci L, Popinigis J, Antosiewicz J. Antioxidant activity of NADH and its analogue--an in vitro study. J Biochem Mol Biol 2004; 37: 416-21. Starkov AA, Fiskum G, Chinopoulos C, Lorenzo BJ, Browne SE, Patel MS, et al. Mitochondrial alpha-ketoglutarate dehydrogenase

18 Current Pharmaceutical Design, 2009, Vol. 15, No. 1

[72]

[73]

[74] [75]

[76]

[77]

[78]

[79]

[80]

[81] [82]

[83]

[84]

[85] [86]

[87]

[88] [89] [90]

[91]

[92]

[93]

[94]

complex generates reactive oxygen species. J Neurosci 2004; 24: 7779-88. Zhang Z, Blake DR, Stevens CR, Kanczler JM, Winyard PG, Symons MC, et al. A reappraisal of xanthine dehydrogenase and oxidase in hypoxic reperfusion injury: the role of NADH as an electron donor. Free Radic Res 1998; 28: 151-64. Daugas E, Susin SA, Zamzami N, Ferri KF, Irinopoulou T, Larochette N, et al. Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis. FASEB J 2000; 14: 729-39. Modjtahedi N, Giordanetto F, Madeo F, Kroemer G. Apoptosisinducing factor: vital and lethal. Trends Cell Biol 2006; 16: 264-72. Lipton SA, Bossy-Wetzel E. Dueling activities of AIF in cell death versus survival: DNA binding and redox activity. Cell 2002; 111: 147-50. Vahsen N, Cande C, Briere JJ, Benit P, Joza N, Larochette N, et al. AIF deficiency compromises oxidative phosphorylation. EMBO J 2004; 23: 4679-89. Hara MR, Thomas B, Cascio MB, Bae BI, Hester LD, Dawson VL, et al. Neuroprotection by pharmacologic blockade of the GAPDH death cascade. Proc Natl Acad Sci USA 2006; 103: 3887-9. Sawa A, Khan AA, Hester LD, Snyder SH. Glyceraldehyde-3phosphate dehydrogenase: nuclear translocation participates in neuronal and nonneuronal cell death. Proc Natl Acad Sci USA 1997; 94: 11669-74. Hara MR, Agrawal N, Kim SF, Cascio MB, Fujimuro M, Ozeki Y, et al. S-nitrosylated GAPDH initiates apoptotic cell death by nuclear translocation following Siah1 binding. Nat Cell Biol 2005; 7: 665-74. Chuang DM, Hough C, Senatorov VV. Glyceraldehyde-3phosphate dehydrogenase, apoptosis, and neurodegenerative diseases. Annu Rev Pharmacol Toxicol 2005; 45: 269-90. Coleman M. Axon degeneration mechanisms: commonality amid diversity. Nat Rev Neurosci 2005; 6: 889-98. Conforti L, Fang G, Beirowski B, Wang MS, Sorci L, Asress S, et al. NAD(+) and axon degeneration revisited: Nmnat1 cannot substitute for Wld(S) to delay Wallerian degeneration. Cell Death Differ 2007; 14: 116-27. Zhai RG, Cao Y, Hiesinger PR, Zhou Y, Mehta SQ, Schulze KL, et al. Drosophila NMNAT maintains neural integrity independent of its NAD synthesis activity. PLoS Biol 2006; 4: e416. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND. Glutathione metabolism and its implications for health. J Nutr 2004; 134: 48992. Arner ES, Holmgren A. Physiological functions of thioredoxin and thioredoxin reductase. Eur J Biochem 2000; 267: 6102-9. Kirkman HN, Gaetani GF. Catalase: a tetrameric enzyme with four tightly bound molecules of NADPH. Proc Natl Acad Sci USA 1984; 81: 4343-7. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007; 87: 245-313. Ushio-Fukai M. Localizing NADPH oxidase-derived ROS. Sci STKE 2006; 2006: re8. Tirone F, Cox JA. NADPH oxidase 5 (NOX5) interacts with and is regulated by calmodulin. FEBS Lett 2007; 581: 1202-8. Inoguchi T, Nawata H. NAD(P)H oxidase activation: a potential target mechanism for diabetic vascular complications, progressive beta-cell dysfunction and metabolic syndrome. Curr Drug Targets 2005; 6: 495-501. Abramov AY, Duchen MR. The role of an astrocytic NADPH oxidase in the neurotoxicity of amyloid beta peptides. Philos Trans R Soc Lond B Biol Sci 2005; 360: 2309-14. Abramov AY, Scorziello A, Duchen MR. Three distinct mechanisms generate oxygen free radicals in neurons and contribute to cell death during anoxia and reoxygenation. J Neurosci 2007; 27: 1129-38. Walder CE, Green SP, Darbonne WC, Mathias J, Rae J, Dinauer MC, et al. Ischemic stroke injury is reduced in mice lacking a functional NADPH oxidase. Stroke 1997; 28: 2252-8. Abramov AY, Canevari L, Duchen MR. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci 2004; 24: 565-75.

Ying et al. [95]

[96]

[97]

[98]

[99]

[100]

[101] [102]

[103]

[104]

[105]

[106]

[107]

[108]

[109]

[110]

[111]

[112]

[113]

[114]

Meischl C, Krijnen PA, Sipkens JA, Cillessen SA, Munoz IG, Okroj M, et al. Ischemia induces nuclear NOX2 expression in cardiomyocytes and subsequently activates apoptosis. Apoptosis 2006; 11: 913-21. Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, et al. Mechanisms of cell death in oxidative stress. Antioxid Redox Signal 2007; 9: 49-89. Ying W, Alano CC, Garnier P, Swanson RA. NAD + as a metabolic link between DNA damage and cell death. J Neurosci Res 2005; 79: 216-23. Zhu K, Swanson RA, Ying W. NADH can enter into astrocytes and block poly(ADP-ribose) polymerase-1-mediated astrocyte death. Neuroreport 2005; 16: 1209-12. Wang S, Xing Z, Vosler PS, Yin H, Li W, Zhang F, et al. Cellular NAD replenishment confers marked neuroprotection against ischemic cell death: role of enhanced DNA repair. Stroke 2008; 39: 2587-95. Ying W, Wei G, Wang D, Wang Q, Tang X, Shi J, et al. Intranasal administration with NAD+ profoundly decreases brain injury in a rat model of transient focal ischemia. Front Biosci 2007; 12: 272834. Ying. W. Therapeutic potential of NAD+ for treating neurological diseases. Future Neurol 2007; 2: 1-4. Kuhn W, Muller T, Winkel R, Danielczik S, Gerstner A, Hacker R, Mattern C, Przuntek H. Parenteral application of NADH in Parkinson's disease: clinical improvement partially due to stimulation of endogenous levodopa biosynthesis. J Neural Transm 1996; 103: 1187-93. Birkmayer JG, Vrecko C, Volc D, Birkmayer W. Nicotinamide adenine dinucleotide (NADH)--a new therapeutic approach to Parkinson's disease. Comparison of oral and parenteral application. Acta Neurol Scand Suppl 1993; 146: 32-5. Demarin V, Podobnik SS, Storga-Tomic D, Kay G. Treatment of Alzheimer's disease with stabilized oral nicotinamide adenine dinucleotide: a randomized, double-blind study. Drugs Exp Clin Res 2004; 30: 27-33. Yang J, Klaidman LK, Nalbandian A, Oliver J, Chang ML, Chan PH, et al. The effects of nicotinamide on energy metabolism following transient focal cerebral ischemia in Wistar rats. Neurosci Lett 2002; 333: 91-4. Mokudai T, Ayoub IA, Sakakibara Y, Lee EJ, Ogilvy CS, Maynard KI. Delayed treatment with nicotinamide (Vitamin B(3)) improves neurological outcome and reduces infarct volume after transient focal cerebral ischemia in Wistar rats. Stroke 2000; 31: 1679-85. Ayoub IA, Lee EJ, Ogilvy CS, Beal MF, Maynard KI. Nicotinamide reduces infarction up to two hours after the onset of permanent focal cerebral ischemia in Wistar rats. Neurosci Lett 1999; 259: 21-4. Ayoub IA, Maynard KI. Therapeutic window for nicotinamide following transient focal cerebral ischemia. Neuroreport 2002; 13: 213-6. Brewer KL, Hardin JS. Neuroprotective effects of nicotinamide after experimental spinal cord injury. Acad Emerg Med 2004; 11: 125-30. Mukherjee SK, Klaidman LK, Yasharel R, Adams JD, Jr. Increased brain NAD prevents neuronal apoptosis in vivo. Eur J Pharmacol 1997; 330: 27-34. Kaneko S, Wang J, Kaneko M, Yiu G, Hurrell JM, Chitnis T, et al. Protecting axonal degeneration by increasing nicotinamide adenine dinucleotide levels in experimental autoimmune encephalomyelitis models. J Neurosci 2006; 26: 9794-804. Chong ZZ, Lin SH, Li F, Maiese K. The sirtuin inhibitor nicotinamide enhances neuronal cell survival during acute anoxic injury through AKT, BAD, PARP, and mitochondrial associated "antiapoptotic" pathways. Curr Neurovasc Res 2005; 2: 271-85. Li F, Chong ZZ, Maiese K. Cell Life versus cell longevity: the mysteries surrounding the NAD+ precursor nicotinamide. Curr Med Chem 2006; 13: 883-95. Klaidman L, Morales M, Kem S, Yang J, Chang ML, Adams JD, Jr. Nicotinamide offers multiple protective mechanisms in stroke as a precursor for NAD+, as a PARP inhibitor and by partial restoration of mitochondrial function. Pharmacology 2003; 69: 150-7.

Roles of NAD + / NADH and NADP+ / NADPH in Cell Death [115]

[116]

[117]

Chong ZZ, Lin SH, Maiese K. The NAD+ precursor nicotinamide governs neuronal survival during oxidative stress through protein kinase B coupled to FOXO3a and mitochondrial membrane potential. J Cereb Blood Flow Metab 2004; 24: 728-43. Chong ZZ, Lin SH, Maiese K. Nicotinamide modulates mitochondrial membrane potential and cysteine protease activity during cerebral vascular endothelial cell injury. J Vasc Res 2002; 39: 13147. Avalos JL, Bever KM, Wolberger C. Mechanism of sirtuin inhibition by nicotinamide: altering the NAD+ cosubstrate specificity of a Sir2 enzyme. Mol Cell 2005; 17: 855-68.

Current Pharmaceutical Design, 2009, Vol. 15, No. 1 [118]

[119]

19

Belenky P, Racette FG, Bogan KL, McClure JM, Smith JS, Brenner C. Nicotinamide riboside promotes Sir2 silencing and extends lifespan via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+. Cell 2007; 129: 473-84. Wang BW, Liao WN, Chang CT, Wang SJ. Facilitation of glutamate release by nicotine involves the activation of a Ca2+/calmodulin signaling pathway in rat prefrontal cortex nerve terminals. Synapse 2006; 59: 491-501.