ANTIOXIDANTS & REDOX SIGNALING Volume 21, Number 1, 2014 ª Mary Ann Liebert, Inc. DOI: 10.1089/ars.2013.5777

FORUM REVIEW ARTICLE

Calcium Signaling Alterations, Oxidative Stress, and Autophagy in Aging Rodrigo Portes Ureshino, Katiucha Karolina Rocha, Guiomar Silva Lopes, Cla´udia Bincoletto, and Soraya Soubhi Smaili

Abstract

Significance: Aging is a multi-factorial process that may be associated with several functional and structural deficits which can evolve into degenerative diseases. In this review, we present data that may depict an expanded view of molecular aging theories, beginning with the idea that reactive oxygen species (ROS) are the major effectors in this process. In addition, we have correlated the importance of autophagy as a neuroprotective mechanism and discussed a link between age-related molecules, Ca2+ signaling, and oxidative stress. Recent Advances: There is evidence suggesting that alterations in Ca2+ homeostasis, including mitochondrial Ca2+ overload and alterations in electron transport chain (ETC) complexes, which increase cell vulnerability, are linked to oxidative stress in aging. As much as Ca2+ signaling is altered in aged cells, excess ROS can be produced due to an ineffective coupling of mitochondrial respiration. Damaged mitochondria might not be removed by the macroautophagic system, which is hampered in aging by lipofuscin accumulation, boosting ROS generation, damaging DNA, and, ultimately, leading to apoptosis. Critical Issues: This process can lead to altered protein expression (such as p53, Sirt1, and IGF-1) and progress to cell death. This cycle can lead to increased cell vulnerability in aging and contribute to an increased susceptibility to degenerative processes. Future Directions: A better understanding of Ca2+ signaling and molecular aging alterations is important for preventing apoptosis in age-related diseases. In addition, caloric restriction, resveratrol and autophagy modulation appear to be predominantly cytoprotective, and further studies of this process are promising in age-related disease therapeutics. Antioxid. Redox Signal. 21, 123–137.

Introduction

T

he aging process is marked by a gradual loss in physical and mental capabilities due to a general decline in organ function, and aging can also be considered a risk factor for cancer, diabetes, and cardiovascular and neurodegenerative diseases. There are numerous aging theories, and most of them are likely compatible and may complement each other. The free radical theory of aging (65) predicts that there is an accumulation of molecules that are damaged by free radicals, mostly by oxidation, and it is extensively reported that there is an increase in oxidative stress in aging (49). In general, senescent cells in culture adopt a flattened, enlarged morphology and exhibit specific molecular markers, such as senescence-associated b-galactosidase, senescence-

associated heterochromatin foci, and the accumulation of lipofuscin granules (91). Although cellular senescence is an adaptive response to stress that contributes to extending the lifespan, this phenomenon might have a negative impact on the survival of the organism. During aging, senescent cells accumulate in proliferative tissues and release various degradative proteases, growth factors, and inflammatory cytokines that compromise the function of nonsenescent neighboring cells, creating a micro-environment which enables preneoplastic cells to develop into tumors, thus counterbalancing longevity with the risk of old-age cancer (183). In this review, we discuss the connection of molecular aging targets with cell physiology, starting from an overview of age-related alterations in Ca2 + signaling and oxidative stress, discussing the autophagy in this process, and developing a

Department of Pharmacology, Federal University of Sa˜o Paulo, Sa˜o Paulo, Brazil.

123

124

link between p53, Sirt1, and IGF-1, while focusing on agerelated alterations in Ca2 + signaling. Calcium, Oxidative Stress, and Apoptosis in Aging

Reactive oxygen species (ROS) are considered the most important oxidant molecules in cells and include superoxide (O2 - ), hydroxyl radical (OH - ), hydrogen peroxide (H2O2), and oxygen singlet (1O2) (158). The largest amounts of ROS are produced in mitochondria through the electron transport chain (ETC) via the leakage of electrons (particularly in complexes I and III) that react with free oxygen in the matrix, producing superoxide (14, 126). These organelles were predicted to participate at the leading edge of aging cellular alterations, because the increase in ROS and other harmful molecules could promote nuclear and mitochondrial DNA (mtDNA) mutations, contributing to a senescent phenotype (125). This outcome is a concern for genetic theories of aging, once DNA mutations cause toxic effects on biological systems (87, 184). Indeed, the accumulation of 8oxoguanosine (a DNA damage marker) in the promoters of genes with decreased expression was observed in the human brain, affecting proteins related to ATP synthesis and vesicle transport (105). Nuclear and mtDNA mutations can affect the transcription of ETC proteins, producing a mismatch in the coupling of mitochondrial respiration and increased ROS generation (66). To strengthen this current hypothesis, Trifunovic and coworkers (173) produced a mouse strain with a defective mtDNA polymerase (PolgA) that had an accumulation of mtDNA mutations associated with a decreased life expectancy and premature aging (displaying weight loss, alopecia, and osteoporosis). During aging, mtDNA mutations are estimated to be present in approximately 1% of the mitochondrial genetic material, which results in a decline in the function of this organelle (102), more frequently found in postmitotic cells (109). Navarro and coworkers (131) demonstrated that there is a 51% reduction in state 3 respiration (ADP-dependent) and 73% and 54% decreases in complex I and IV (ETC) activity, respectively, in the hippocampus of aged rats, in accordance with the previously reported reduction of the protein expression of these complexes (109). Besides, mitochondrial dynamics, particularly fusion and fission, are important to the maintenance of mtDNA integrity (11, 150). During these processes, mediated by proteins such as mitofusins (Mfs1 and Mfs2), Opa1 (fusion), and Drp1 (fission), there is a combination of mutated and normal mtDNA, known as mtDNA heteroplasmy. The excess of fission can cause respiratory impairment and an increase in oxidative stress (191); if fusion prevails, the presence of large and defective mitochondria can hamper mitochondrial autophagy (55) (described in section ‘‘Autophagy in Aging and Neurodegeneration’’), which may play an important role in cellular protection during senescence (95). Several authors have reported that there is reduced antioxidant capacity in aging (80, 154). Tian and coworkers (169) found a large amount of protein carbonyls but no change in lipid peroxidation in the heart, liver, kidney, and brain of aged rats; the latter also showed a reduction in the activity of catalase and no changes in superoxide dismutase (SOD) and glutathione peroxidase (GPx). The peroxisomes, single membrane-bound organelles containing anti-oxidants and

URESHINO ET AL.

oxidases (116, 178), play an important role in the aging process (137, 166). Several authors have reported alterations in catalase, acyl-CoA oxidase, and b-oxidation in the liver of Wistar rats (8), with an increase in b-oxidation in the aged brain (13). In fact, it was observed that systemic catalase overexpression in mitochondria extends the lifespan of mice (149). Ca2 + modulates the ETC and ATP production by its activity in dehydrogenases of the tricarboxylic acid cycle (59), and ATP synthase complex activation (167), in addition to its importance in maintaining the mitochondrial membrane potential (DJm) (38). Lu and coworkers (105) analyzed the expression of several genes in aged human brains and found, among other differences, alterations in the expression of Ca2 +-related genes. Accordingly, it has been proposed that a sudden imbalance in Ca2 + signaling can be considered an additional factor for cellular demise during aging (83), because it is mandatory for mitochondrial respiratory function. There are extensive data linking alterations in Ca2 + signaling, mitochondria, and apoptosis (39, 133). In particular, in the central nervous system (CNS), excessive glutamate in synaptic terminals can lead to Ca2 + overload (50, 132) via the overactivation of ionotropic NMDA (N-methyl-D-aspartic acid) receptors (140), which is called excitotoxicity. This phenomenon can contribute to cell death that occurs during brain aging (22, 177) and neurodegenerative processes (7, 30, 35). Despite excitotoxicity also occurring in glial cells, neurons are more susceptible to cell injury caused by glutamate. Astrocytes are known to support neurons (182) and to protect against oxidative stress and excitotoxicity (40). They are also pivotal in the neurotransmission process. Furthermore, GFAP, an astrocyte marker, is overexpressed in CNS of aged rats (48, 122); Castillo-Ruiz and coworkers observed an early astrogliosis after NMDA infusion in the cortex and striatum of aged rats, accompanied by increased degenerated neurons (25), showing that neurons are more vulnerable to injuries than glial cells. Mitochondria are particularly involved in the excitotoxicity process (for a review, see Smaili et al., 2011) (157). Ca2 + is taken up by mitochondria through a uniporter. This electrogenic channel has a low affinity for Ca2 + and forms ‘‘microdomains’’ of Ca2 + within the endoplasmic reticulum (ER), which releases Ca2 + through inositol trisphosphate receptor (IP3R) and ryanodine receptors (RyR) (143). The massive Ca2 + increase evoked by glutamate can cause mitochondrial Ca2 + overload (79, 136). Ca2 + accumulation in the mitochondria drives unregulated bioenergetics, which, in turn, can generate more ROS and eventually open the permeability transition pore (PTP) (5, 181), reducing cell viability and, ultimately, leading to cell death. This pathway is illustrated in Figure 1. Studies combining patch voltage clamping and fluorescence microscopy in neurons showed that intracellular Ca2 + buffering occurs in two steps: fast and slow (127). (i) The first stage, which appears to have high activity in aging, likely corresponds to a compensatory mechanism against a large influx of ions from voltage-operated Ca2 + channels (VOCC), and intracellular Ca2 + storage may have an important role in this process. Kirischuk and Verkhatsky (86) reported a decline in caffeine-sensitive ER storage in the cerebellar granule neurons of aged animals, and caffeine produced a plateau that was significantly higher in a cell senescence model (29). Indeed, it was demonstrated that the ER can store more Ca2 + in the striatum of aged rats (177). (ii) The second

CALCIUM, ROS, AUTOPHAGY, AND AGING

125

FIG. 1. Excitotoxicity, ROS, and apoptosis. On excessive glutamate (Glu) stimulation, there is a massive Ca2 + increase due to an influx by ionotropic (iGluR; e.g., NMDA) and metabotropic receptors (mGluR group I), which activates Gq protein, stimulates phosphatidylinositol 4,5-bisphosphate cleavage, resulting in IP3 formation and activation of IP3R in the ER. This release of Ca2 + from the ER can be enhanced by the activation of Ca2 + -induced Ca2 + release via RyR. In mitochondria, Ca2 + primarily enters through the uniporter and activates the tricarboxylic acid cycle (TCA) and ATP synthase (ATPs), prompting the ETC and elevating DJm. This process enhances the probability of electron leakage, particularly via complexes I and III; these electrons then react with free oxygen in the matrix and produce superoxide. This process is the origin of ROS, which can damage cell membranes and DNA, evoking DDR (DNA damage response) and enhancing and activating p53 through its phosphorylation (p) and acetylation (a). p53 directly (by transactivation in the cytosol) and indirectly (by transcription in the nucleus) activates Bax and other pro-apoptotic proteins. Bax can oligomerize at the mitochondrial outer membrane, releasing cytochrome c and APAF-1; in the presence of pro-caspase 9 and dATP, the apoptosome is formed, which activates the intrinsic apoptosis pathway. p53 also modulates oxidative stress by inducing the expression of several antioxidants (e.g., Sesn, Gpx, and MnSod) and mitochondrial respiration modulators (e.g., Tigar, Sco2). ([: stimulation) (>: inhibition). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars stage, corresponding to slow Ca2 + buffering, appears to be more dependent on mitochondria (68). Lopes and coworkers (104) found that there was a greater release of mitochondrial Ca2 + after protonophore (FCCP - carbonyl cyanide 4(trifluoromethoxy)phenylhydrazone) addition in the colon of aged rats. In addition, it was demonstrated that the aged striatum had increased mitochondrial Ca2 + storage in the CNS, which was accompanied by an increase in ROS production and a reduction in DJm (177). In summary, mitochondrial Ca2 + accumulation might promote an increase in ROS and alter bioenergetics, leading to PTP

opening and a loss of DJm (64), which can cause apoptosis (155, 156). The link between apoptotic proteins of the Bcl-2 family, which comprises pro- (e.g., Bax, Bak) and antiapoptotic (e.g., Bcl-2, Bcl-xl) members, and Ca2 + signaling has been extensively studied. In the ER, Bcl-2 was reported to interact with IP3R and reduce the release of Ca2 + (145), though some studies have indicated that Bcl-2 could increase (56), reduce (46, 70, 138), or have no effect (187) on the ER Ca2 + content. In mitochondria, Bcl-2 overexpression increases the concentration or accumulation capacity of Ca2 + (70, 90, 199),

126

URESHINO ET AL.

FIG. 2. PC12 cells overexpressing Bcl-2 (Bcl-21) display alterations in intracellular Ca21 stores. In the left panel, Bcl-2 + cells show a reduced ER Ca2 + content, which was evaluated after the incubation with the ER Ca2 + mobilizer agent Tapsigargin (Tap). On the other hand, the overexpression of Bcl-2 increases mitochondrial Ca2 +, which was analyzed after the addition of the mitochondrial Ca2 + mobilizer agent FCCP. *Significant difference (p £ 0.05, Student’s t test). Adapted from Hirata et al., 2012 (70). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

which is associated with the elevation of DJm and the mitochondrial volume (90) (Fig. 2). The proapoptotic protein Bax otherwise induces ER and mitochondrial Ca2 + release and the loss of DJm, which might be related to ETC inhibition and the partial release of cytochrome c (24, 155) (Fig. 3). Alterations in the expression of Bcl-2 can be directly related to oxidative stress, because Bcl-2 modulates the intracellular levels of ROS (90). Conversely, an increase in ROS can lead to a reduction in Bcl-2 levels (69). Indeed, greater ROS production was accompanied by a reduction in Bcl-2 expression in the striatum of aged rats (177), but with an enhanced activity of the antioxidant enzyme GPx, resulting in no change in lipid peroxidation (Fig. 4).

An increase in the Bax/Bcl-2 protein ratio may have a positive correlation with apoptosis, and an increase in the bax/bcl-2 mRNA ratio may also indicate an increase in the number of apoptotic cells in aging brains (37, 168, 177). Proteins such as Bcl-2 and Bax share a common C-terminal hydrophobic domain that enables intimate contact with organelles, including the ER, mitochondria, and nucleus (196). Briefly, pro-apoptotic proteins bind to the membranes of organelles (such as mitochondria) and promote the release of proapoptotic factors, such as cytochrome c (73, 155). In the cytoplasm, cytochrome c forms a complex (apoptosome) with APAF-1/caspase-9 (157), and caspase-9 cleaves procaspase-3, activating it and subsequently cleaving inhibitor

FIG. 3. Bax induces Ca21 mobilization from intracellular stores and releases cytochrome c, in primary culture astrocytes. (A) Recombinant Bax evokes Ca2 + rise even after the addition of Tapsigargin (Tap) or FCCP. (B) In addition, the recombinant Bax induces the release of cytochrome c from mitochondria to the cytosol. *Significant difference (p £ 0.05, Student’s t test). Adapted from Carvalho et al., 2004 (24). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

CALCIUM, ROS, AUTOPHAGY, AND AGING

127

FIG. 4. In aging, the increased generation of oxidant molecules (e.g., ROS) can be balanced by the increase of antioxidant enzymes (e.g., Gpx), which may result in no changes in lipid peroxidation in aged striatum. *Significant difference (p £ 0.05, Student’s t test). Adapted from Ureshino et al., 2010 (177).

of caspase-activated DNase (iCAD), which releases CAD to fragment the DNA (42). There is also a concomitant release of endonuclease G (EndoG), which also participates in DNA fragmentation (162), a hallmark of apoptosis (82). Autophagy in Aging and Neurodegeneration

Autophagy, which literally means ‘‘self-eating’’ and was coined by Nobel Laureate Christian de Duve in 1963, enables cells to digest cytosolic components via lysosomal degradation. In addition to scavenging the cytosol for macromolecules and damaged organelles, the process of autophagy also provides a way to supply cells with amino acids and energy due to recycling (88). With regard to the turnover of longlived proteins and the removal of damaged organelles and cellular debris, autophagy is believed to constitute an antiaging process (9). During aging, particularly in tissues populated by postmitotic cells, there is an accumulation of insoluble particles

FIG. 5. Autophagic process in aged cells. During aging, particularly in post mitotic cells, there is a progressive accumulation of lipofuscin, which impairs the autophagy of toxic protein aggregates and mitophagy. Then, damaged mitochondria are not removed in aged cells, which increase oxidative stress. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

called lipofuscins, which can aggregate in autophagosomes and hamper the autophagic system (18, 91) (Fig. 5), a valuable protective mechanism for the cell (110). Since the autophagic system is impaired in aging, damaged mitochondria produce a large amount of ROS and cannot be recycled, which increases cell oxidative stress (18). In addition, analyses of brain gene expression (i.e., Atg5, Atg7, and Beclin 1) in the elderly revealed a down-regulation of autophagic genes compared with young subjects; conversely, an up-regulation was observed in patients with Alzheimer’s disease (47). In 1935, McCay and coworkers reported that reducing caloric intake in rats by 20%–40% extends the lifespan (111, 117), and caloric restriction (CR) was also found to extend the lifespan in yeast, worms, rodents, and monkeys (153). In addition, it is well described that autophagy may be important for increasing longevity (67, 77). Moreover, CR modulates multiple genetic pathways that are involved in aging, including protein homeostasis, which is primarily maintained by removing damaged proteins through the ubiquitin– proteasome system, and autophagy, a process that protects cells from undesirable effects produced by damaged proteins (101), as previously mentioned. From another perspective, a reduction in mitophagy (mitochondrial autophagy) may also be involved in aging-related diseases, such as Parkinson’s disease. The etiology focuses on genetic and environmental factors (51) and is associated with aging, though there are many cases of the early onset of this disease (54). In this process, the protein aggregation of filamentous intracytoplasmic inclusions called Lewy bodies cannot be removed by autophagy (183). Approximately 5% of Parkinson’s disease cases are caused by genetic changes that lead to alterations in the expression of proteins such as Pten-induced novel kinase 1 (PINK1) and parkin (52, 179), which are related to mitophagy (194). When mitochondria are damaged, PINK1 accumulates in the mitochondrial outer membrane for the recruitment of parkin and subsequent mitochondrial degradation (130), and one of the most common stimuli used to sensitize the PINK1-parkin system is the dissipation of DJm (114). It is interesting that there is a reduction in DJm in the cells of many tissues during aging (60, 177, 190). Taken together, the intersection between oxidative stress, Ca2 + , and autophagy should be clarified with regard to molecular alterations in the aging process. Molecular Aging Targets

As mentioned earlier, some proteins are potential molecular targets for improving lifespan or at least reducing the age-associated risk for degenerative diseases. Proteins such

128

FIG. 6. Relationship between altered age-related molecules. IGF-1 signaling inhibits the transcriptional activity of FoxO3, which modulates p53 through the up-regulation of p19Arf. Sirt1 participates in the deacetylation of FoxO3 and p53, modulating their transcriptional functions. ([: stimulation) (>: inhibition).

as the transcription factor forkhead box protein O3 (FoxO3), whose polymorphisms have been associated with extreme longevity in humans (3, 189), and other molecules remain the focus of orthomolecular therapeutics for aging-associated diseases. FoxO3, a transcriptional activator that modulates DNA repair and apoptosis (16, 141, 192), plays a role in oxidative stress by causing a reduction in the expression of mitochondrial respiratory complexes and, thus, in mitochondrial respiratory activity (45). In addition, FoxO3 acts in the modulation of SOD2 (89), catalase (164), peroxiredoxin III (28), and sestrin 3 (26, 61), though the role of FoxO3 in Ca2 + signaling still needs to be established. The following molecules have been associated with FoxO3. (i) p53: FoxO3 leads to the activation of p53-dependent apoptosis (193) and up-regulates p19Arf, an upstream regulator of p53 (12). (ii) Sirt1: FoxO3 and p53 are deacetylated by the Sirt1 deacetylase (17, 124). (iii) IGF-1: insulin and growth factors inhibit the transcriptional activity of FoxO3 by phosphorylating its nuclear export sequence (16) (Fig. 6). These proteins are very important modulators of oxidative stress and autophagy and

URESHINO ET AL.

play a considerable role in Ca2 + signaling. Next, we discuss the relationship between age-related molecules and the critical alterations in cell physiology. p53 is a tumor suppressor protein that is known to induce apoptosis via the up-regulation of proteins at the transcriptional level or via cytosolic action (57). The most important activity of p53 is an increase in Bax expression (121) and the transactivation of Bax in the cytosol (27), leading to the oligomerization of Bax at the mitochondrial outer membrane to release pro-apoptotic factors. The p53 DNA-binding domain is responsible for its transcriptional activity and is also important in the cytosol, where it interacts with Bcl-2, acting similar to a BH3-only protein (119), potentially switching the cell fate to apoptosis. With regard to the aging process, several groups have used genetically modified animal models by mutating or overexpressing p53, and such mice showed an increased capacity for tumor resistance but a reduced (174), unaltered (53), or increased lifespan (112), depending on p53 activation. In the striatum of aged rats, there is an increase of phosphorylated p53, which can increase bax gene expression (Fig. 7). The cellular alterations that underlie the activation of p53 remain under discussion and may be associated with oxidative stress and Ca2 + homeostasis. Jorda´n and coworkers (78) showed that p53 is important for the hippocampal neuronal cell death promoted by the depolarization agent veratridine, which induces Ca2 + overload (Fig. 8). On exposure to UV light, a classic p53 inducer (93), there is a differential modulation of mitochondrial uniporter activity in young and aged striatum slices, which may be dependent on p53 only in aged rats (Ureshino and coworkers, unpublished data). Notably, the Ca2 + -binding protein S100B is capable of down-regulating p53 in melanoma cells, thereby preventing apoptosis (100). However, the exact relationship between p53 and Ca2 + remains to be established. With regard to oxidative stress, it has been shown that apoptosis caused by p53 activation is dependent on the previous elevation of ROS, leading to the activation of the intrinsic pathway of apoptosis (139). Similarly, it has been demonstrated that the activation of p53 requires an exogenous source of ROS to cause apoptosis; in fact, cell death by oxidative stress is prevented in the absence of p53 (106). As previously reported, oxidative stress in aging promotes the accumulation of DNA damage (approximately two times higher compared with young animals) (2). Dorszewka and Adamczewska-Goncerzewicz (36) found an increase in p53

FIG. 7. Increase in phosphorilated p53 (p.p53) expression is related to a rise in bax gene expression in aged striatum, which may cause an increase in cell vulnerability to apoptosis. *Significant difference (p £ 0.05, Student’s t test).

CALCIUM, ROS, AUTOPHAGY, AND AGING

129

FIG. 8. Interrelationship between Ca21 signaling, ROS generation, autophagy, and some important molecules in the aging process. Molecules such as Sirt1, p53, and IGF-1 can modulate autophagy and cross-talk with Ca2 + signaling and oxidative stress. Sirt1 up-regulates autophagy by deacetilating ATG5 and ATG7, helps in LC3 II conversion, and activates AMPK, which inhibits mTOR. Caloric restriction (CR) also stimulates AMPK through an increase in the AMP/ATP ratio or NAD + /NADH and promotes Sirt1 expression. Resveratrol acts directly and increases Sirt1 and ROS scavenger. Sirt1 modulates Ca2 + signaling, acting on SERCA and CaM. p53 increases sestrin (Sesn) levels, which participates in the peroxiredoxin cycle, reducing ROS levels, in addition to the stimulation of AMPK. Although the mechanism is not clear, cytosolic p53 acts by inhibiting autophagy and also modulates Ca2 + by increasing VOCC function. In contrast, the IGF-1 pathway has a pro-aging activity, enhancing ROS production and reducing GPx. IGF-1/IGFR binding increases PI3K activity through insulin receptor substrate 1 (IRS-1), elevating IP3 levels (releasing Ca2 + from the ER) and activating AKT, resulting in the activation of mTOR (through the inhibition of TSC2 and Rheb) and the inhibition of autophagy. Finally, the elevation of ROS can contribute to the p53-mediated cell death ([: stimulation) (>: inhibition). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

mRNA in the cortex, cerebellum, and spinal cord of 24month-old rats, with only the cortex exhibiting a significant increase in p53 protein levels, which was associated with a higher Bax/Bcl-2 ratio and DNA fragmentation. It should be noted that p53 can regulate redox signaling (148) via the expression of antioxidant enzymes such as GPx (163) and manganese superoxide dismutase (MnSOD) (75). p53 was recently shown to be intimately involved in oxidative metabolism, as it up-regulates the expression of proteins called sestrins (sestrin 1 and sestrin 2) (20), which participate in the ROS depuration cycle of peroxiredoxin, reducing this enzyme from its SO2H to its SOH form (Fig. 8). In addition, some authors have reported that p53 can modulate biochemical processes, including glycolysis and mitochondrial oxidative phosphorylation, through the expression of genes encoding the glycolysis regulatory protein TIGAR (TP53induced glycolysis regulator) (107) or complex IV protein of the ETC, such as SCO2 (synthesis of cytochrome c oxidase 2) (113), thereby controlling the oxidative stress status. Moreover, p53 is also related to the modulation of autophagy, as it can transactivate several proteins (for a review, see Maiuri et al., 2010) (108). Damage-regulated autophagy modulator, a phylogenetical lysosomal protein (31), the b1 and b2 subunits of AMPK (adenosine monophosphateactivated protein kinase) (44), and sestrin 1 and sestrin 2 (19), which also activate AMPK, are direct targets of p53. AMPK activation is responsible for inhibiting the mammalian target of rapamycin (mTOR), promoting autophagy (146). De-

spite an unclear mechanism, it was reported that p53 can also inhibit autophagy when it is present in the cytosol (165) (Fig. 8). Sirtuins (Sirt) are nicotinamide adenine dinucleotide (NAD + )-dependent class III histone deacetylases that function in both the nucleus and cytoplasm (63) and are involved in several biological processes, such as oxidative metabolism, inflammation, DNA transcription, insulin secretion, cellular senescence, apoptosis, and DNA repair (62, 72). Several studies support the importance of Sirt1 in the aging process, and it has been noted that Sirt1 and its yeast homolog Sir2 play an important role in lifespan (72). According to Michain and Sinclair (118), the overexpression of Sir2 prolonged longevity in yeast, flies, and worms. Sirt1 is the most studied member of the family, because it has been shown to prevent oxidative stress and DNA damage (62). Haigis and Sinclair (63) discussed that the anti-aging effects of Sirt1 could be related to increased stress resistance in mammalian cells in vivo, as Sirt1 accumulation has been associated with stress responses (185, 195). It was demonstrated that an Sirt1 agonist restored the age-related loss of cardioprotection (171), and the overexpression of this protein in pancreatic b cells in mice caused an enhancement of glucosestimulated insulin secretion (10). Despite the abundant evidence of the anti-aging role of this protein, some authors have provided data regarding neuroprotection on Sirt1 inhibition (99) and have reported that Sirt1 in Caenorhabditis elegans and Sir2 can also accelerate cellular aging and death (43,103),

130

thus constituting an issue under continuous discussion in the aging process. Sirt1 regulates the cellular metabolic state through the phosphorylation of AMPK (92), which is activated by an elevation in the AMP/ATP ratio or certain kinases, such as liver kinase B1 (LKB1) and Ca2 + /calmodulin-dependent protein kinase kinase b (CaMKKb) (71). Sirt1 activation and NAD + increases are associated with an increase in the intracellular Ca2 + concentration (134) and can also activate Ca + 2 channels, promote a vasorelaxant effect (176), and improve SERCA2a (sarco-ER Ca2 + -ATPase, an ER Ca + 2reuptake channel) expression and Ca + 2 uptake in the myocardium (161) (Fig. 8). CR has been reported to increase Sirt1 expression and promote autophagy through the AMPK pathway (23), which inhibits mTOR (146). Furthermore, Sirt1 is involved in the autophagy process through the deacetylation of ATG 5 and ATG 7; aids in autophagosome formation (94); and is also associated with the conversion of LC3I to LC3II via proteolytic cleavage and lipidation (123) (Fig. 8). CR also promotes the stimulation of neurogenesis and increases synaptic plasticity, resulting in the resistance to cognitive decline (115). Resveratrol (3-5-4¢ trihydroxystilbene), a polyphenol present in various vegetables and fruits, has been reported to be a powerful antioxidant and protector of mitochondria against oxidative stress (120) through its activity as a free radical scavenger (1). Resveratrol was also demonstrated to protect mitochondria via an increase in citrate synthase activity and to improve mitochondrial respiration in the presence of fatty acid substrates (170). It has been reported that resveratrol acts directly or indirectly on the activation of Sirt1, enhancing its expression (63); similar to CR, resveratrol has been associated with increased autophagy (123). Insulin-like growth factors (IGFs) are anabolic and mitogenic hormones that regulate cell growth and differentiation, cell metabolism, and apoptosis (96, 142). IGF-1 stimulates the IGF-1 receptor (IGFR) and activates insulin receptor substrate 1 (IRS-1), the major cytosolic substrate (129, 147), leading to the activation of phosphoinositide 3-kinase (PI3K) (128, 147), which targets AKT (198). In the autophagic pathway, AKT inhibits tuberous sclerosis protein 2 (TSC2); the heterodimer TSC1/2 antagonizes the mTOR signaling pathway via the inhibition of Rheb (Ras homologue enriched in the brain). Thus, AKT activation leads to the activation of mTOR and blocks autophagy (98). In addition, active PI3K increases the levels of IP3 (76, 147), which causes Ca2 + release from the ER; this Ca2 + is, consequently, taken up by mitochondria, thereby elevating mitochondrial respiration in cardiomyocytes (21, 76). This pathway is shown in Figure 8. Moreover, IGF-1 was also correlated with increased L-type Ca2 + channel gene expression in skeletal muscle (186), enhancing the amplitude of the Ca2 + current but not modifying the voltage dependence of Ca2 + channel currents in the cortex of young and aged rats (151). Since IGF-1 activates cell growth and survival, its synthesis is down-regulated in the aging process (74), and there is evidence that associates the reduction in circulating growth hormone (GH) and IGF-1 with age-related functional deficits in many organs (84, 159). This situation is evident in the cardiac system where IGF-1 acts as an important protector in the heart (175), as the activation of IGFR promotes the

URESHINO ET AL.

conservation of telomere length and the growth of cardiac progenitor cells (172). Furthermore, the overexpression of IGF-1 has been related to cardiomyocyte contraction improvement in aged mice (97), and the loss of IGF-1 contributes to an age-related decline in cardiovascular function (34). Dardevet and coworkers (33) reported a reduction in the IGF-1 response in aged rats due to the diminished expression (*80%) of IGFR and consequent reduced glucose transport. IGF-1 may modulate oxidative metabolism either by enhancing ROS production (144) or by reducing certain antioxidant enzymes, such as GPx (15) (Fig. 8). Guevara-Aguirre and coworkers (58) monitored individuals with GH receptor mutations, which are related to IGF-1 deficiency, extracted their serum and treated cells, and observed a reduction in the expression of RAS, PKA (protein kinase A), and TOR and the up-regulation of SOD2. Under physiological conditions, Sirt1 can increase IGF-1 signaling, promote ROS production, and alter redox signaling (99). In addition, resveratrol, an Sirt1 activator, have been associated with a reduction in insulin/IGF-1 signaling and increased mitochondrial biogenesis (6, 197), thus promoting protection against age-related alterations. Although a reduction in IGF-1 signaling appears to be crucial for increasing longevity, the association between IGF-1 and sirtuins remains controversial (43, 72, 99, 103, 197). The conserved insulin/IGF-1 signaling pathway regulates autophagy through mTOR by activating AKT (98, 146), and this interaction controls cell metabolism and longevity in various species (146). In mammals and worms, a loss of function in the insulin/IGF-1 signaling pathway inhibits tumor growth by activating the p53-mediated cell death pathway (180). IGF-1 induces premature cellular senescence by inhibiting sirtuins and activating p53 (85), and it is also related to the activation of mTOR (188). Thus, a reduction in insulin/IGF-1 signaling increases the lifespan of C. elegans (81) and mice (4). This repression can be related to the activation of AMPK (41), which can also reverse age-related alterations (32). Interestingly, mutations that reduce the activity of the IGF-1 receptor (160) and also in IGF-1 pathway genes (135) were found in centenarians. The mouse strains Ames dwarf, Snell dwarf, GHRKO, and Little mice are known for their reduced body size and increased longevity. These strains share a GH deficiency, and the proposed mechanism of delayed aging included reduced systemic IGF1 levels (for a review, see Bartke and Brown-Borg, 2004) (4). It is interesting that Ames dwarf mice display mTOR downregulation, additional evidence that autophagy increases longevity (152). Figure 8 summarizes the relationship mentioned earlier between molecular aging targets (p53, Sirt1, and IGF-1) and the critical age-related alterations in cell physiology, including Ca2 + signaling, oxidative stress, and autophagy. Concluding Remarks

The underlying causes of aging and the molecular mechanisms of longevity might help in preventing age-associated diseases and/or improving the quality of life. Although previous studies of aging were primarily observational, describing changes in the morphology and function of tissues, organs, and organisms, more recent in vitro and in vivo studies involving age models have identified relevant molecules and their respective signaling pathways.

CALCIUM, ROS, AUTOPHAGY, AND AGING

Within the scope of this review, Ca2 + signaling may be associated with certain apoptotic pathways; however, despite being the focus of many studies as an important element for cell death, the exact role of Ca2 + signaling in this process is unknown. Nonetheless, it is known that Ca2 + actively participates in cell death after excitotoxic stimulation. In this case, the increased influx of Ca2 + primarily through NMDAR leads to the accumulation of Ca2 + in mitochondria, which might accelerate oxidative phosphorylation and modify mitochondrial bioenergetics, contributing to an increase in ROS production. This event may contribute to the occurrence of PTP opening and the release of proapoptotic factors. In fact, alterations in some proteins such as p53, Sirt1, and IGF-1, which may change signaling, can be related to an increased susceptibility to Ca2 + -mediated cell death. Meanwhile, a direct link between the molecules, Ca2 +signaling, and aging remains to be clarified. In addition, since the role of autophagy within many cellular contexts appears to be predominantly cytoprotective and the induction of autophagy by pharmacological or genetic methods has life-extending effects, further studies of this fundamental cellular process are necessary to better address its role in the prevention of aging effects and/or treatment of age-related degenerative diseases. This study highlights the complexity of oxidative stress, Ca2 + signaling, and autophagy interaction with age-related interest molecules and underscores the importance of understanding the molecular mechanisms through which these processes of autophagy function in aging. This relation is not fully established, and there are some controversial studies that keep this issue under discussion. Finally, further investigations between molecular aging theories and cellular metabolism may promote advances in therapeutics and increases in healthspan. Acknowledgments

This work was supported by Fundac¸a˜o de Amparo a` Pesquisa do Estado de Sa˜o Paulo (FAPESP), Conselho Nacional de Desenvolvimento Cientı´fico e Tecnolo´gico (CNPq), and Coordenac¸a˜o de Aperfeic¸oamento de Pessoal de Nı´vel Superior (CAPES). References

1. Alarco´n de Lastra C and Villegas I. Resveratrol as an antiinflammatory and anti-aging agent: Mechanisms and clinical implications. Mol Nutr Food Res 49: 405–430, 2005. 2. Ames BN, Shigenaga MK, and Hagen TM. Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci U S A 90: 7915–7922, 1993. 3. Anselmi CV, Malovini A, Roncarati R, Novelli V, Villa F, Condorelli G, Bellazi R, and Puca AA. Association of the FOXO3A locus with extreme longevity in a southern Italian centenarian study. Rejuvenation Res 12: 95–104, 2009. 4. Bartke A and Brown-Borg H. Life extension in the dwarf mouse. Curr Top Dev Biol 63: 189–225, 2004. 5. Basso E, Fante L, Fowlkes J, Petronilli V, Fortes MA, and Bernardi P. Properties of the permeability transition pore in mitochondria devoid of cyclosporin D. J Biol Chem 280: 18558–18561, 2005. 6. Baur JA and Sinclair DA. Therapeutic of resveratrol: the in vivo evidence. Nat Rev Discov 5: 493–506, 2006.

131

7. Beal MF. Excitotoxicity and nitric oxide in Parkinson’s disease pathogenesis. Ann Neurol 44: S110–S114, 1998. 8. Beier K, Volkl A, and Fahimi HD. The impact of aging on enzyme proteins of rat liver peroxisomes: quantitative analysis by immunoblotting and immunoelectron microscopy. Virchows Arch B Cell Pathol 63: 139–146, 1993. 9. Bergamini E. Autophagy: a cell repair mechanism that retards ageing and age-associated diseases and can be intensified pharmacologically. Mol Aspects Med 27: 403– 410, 2006. 10. Bordone L, Motta MC, Picard F, Robinson A, Jhala US, Apfeld J, McDonagh T, Lemieux M, McBurney M, Szilvasi A, Easlon EJ, Lin SJ, and Guarente L. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol 4: e31, 2006. 11. Bossy-Wetzel E, Barsoum MJ, Godzik A, Schwarzenbacher R, and Lipton SA. Mitochondrial fission in apoptosis, neurodegeneration and aging. Curr Opin Cell Biol 15: 706–716, 2003. 12. Bouchard C, Lee S, Paulus-Hock V, Loddenkemper C, Eilers M, and Schmitt CA. FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf. Genes Dev 21: 2775–2787, 2007. 13. Bourre JM and Piciotti M. Alterations in eighteen-carbon saturated, monounsaturated and polyunsaturated fatty acid peroxisomal oxidation in mouse brain during development and aging. Biochem Mol Biol Int 41: 461–468, 1997. 14. Boveris A, Cadenas E, and Stoppani AO. Role of ubiquinone in the mitochondrial generation of hydrogen peroxide. Biochem J 156: 435–444, 1976. 15. Brown-Borg HM, Bode AM, and Bartke A. Antioxidative mechanisms and plasma growth hormone levels: potential relationship in the aging process. Endocrinol 11: 41–48, 1999. 16. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, Blenis J, and Greenberg ME. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868, 1999. 17. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu LS, Cheng H, Jedrychowski MP, Gygi SP, Sinclair DA, Alt FW, and Greenberg ME. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303: 2011–2015, 2004. 18. Bru¨nk UT and Terman A. The mitochondrial-lysosomal axis theory of aging: accumulation of damaged mitochondria as a result of imperfect autophagocytosis. Eur J Biochem 269: 1996–2002, 2002. 19. Budanov AV and Karin M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 134: 451–460, 2008. 20. Budanov AV, Sablina AA, Feinstein E, Koonin EV, and Chumakov PM. Regeneration of peroxiredoxins by p53regulated sestrins, homologs of bacterial AhpD. Science 304: 596–600, 2004. 21. Cadenas C, Miller RA, Smith I, Bui T, Molgo J, Muller M, Vai H, Cheung K, Yang J, Parker I, Thompson CB, Birnbaum MJ, Hallous KR, and Foskett JK. Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca + transfer to mitochondria. Cell 142: 270–283, 2010. 22. Cady C, Evans MS, and Brewer GJ. Age-related differences in NMDA responses in cultured rat hippocampal neurons. Brain Res 921: 1–11, 2001.

132

23. Canto´ C, Jiang LQ, Deshmukh AS, Mataki C, Coste A, Logouge M, Zierath JR, and Auwerx J. Interdependence of AMPK and Sirt 1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab 11: 213–219, 2010. 24. Carvalho AC, Sharpe J, Rosenstock TR, Teles AF, Youle RJ, and Smaili SS. Bax affects intracellular Ca2 + stores and induces Ca2 + wave propagation. Cell Death Differ 11: 1265–1276, 2004. 25. Castillo-Ruiz MM, Campuzano O, Acarin L, Castellano B, and Gonzalez B. Delayed neurodegeneration and early astrogliosis after excitotoxicity to the aged brain. Exp Geront 42: 343–354, 2007. 26. Chen CC, Jeon SM, Bhaskar PT, Nogueira V, Sundararajan D, Tonic I, Park Y, and Hay N. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev Cell 18: 592–604, 2010. 27. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD, Schuler M, and Green DR. Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science 303: 1010–1014, 2004. 28. Chiribau CB, Cheng L, Cucoranu IC, Yu YS, Clempus RE, and Sorescu D. FOXO3A regulates peroxiredoxin III expression in human cardiac fibroblasts. J Biol Chem 283: 8211–8217, 2008. 29. Clodfelter GV, Porter NM, Landfield W, and Thibault O. Sustained Ca2 + -induced Ca2 + -release underlies the postglutamate lethal Ca2 + plateau in older cultured hippocampal neurons. Eur J Pharmacol 447: 189–200, 2002. 30. Coyle JT and Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science 262: 689–695, 1993. 31. Crighton D, Wilkinson S, O’Prey J, Syed N, Smith P, Harrison PR, Gasco M, Garrone O, Crook T, and Ryan KM. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 126: 121–134, 2006. 32. Cuervo AM. Autophagy and aging: keeping that old broom working. Trends Genet 24: 604–612, 2008. 33. Dardevet D, Sornet C, Attaix D, Baracos VE, and Grizard J. Insulin-like growth factor-1 and insulin resistance in skeletal muscles of adult and old rats. Endocrinol 134: 1475–1484, 1994. 34. Delaughter MC, Taffet GE, Fiorotto ML, Entman ML, and Schwartz RJ. Local insulin-like growth factor I expression induces physiologic, then pathologic, cardiac hypertrophy in transgenic mice. FASEB J 13: 1923–1929, 1999. 35. Difiglia M. Excitotoxic injury of the neostriatum: a model for Huntington’s disease. Trends Neurosci 13: 286–289, 1990. 36. Dorszewska J and Adamczewska-Goncerzewicz Z. Oxidative damage to DNA, p53 gene expression and p53 protein level in the process of aging in rat brain. Resp Physiol Neurobiol 139: 227–236, 2004. 37. Dorszewska J, Adamczewska-Goncerzewicz Z, and Szczech J. Apoptotic proteins in the course of aging of central nervous system in the rat. Respir Physiol Neurobiol 139: 145–155, 2004. 38. Duchen MR. Ca(2 + )-dependent changes in the mitochondrial energetics in single dissociated mouse sensory neurons. Biochem J 283: 41–50, 1992. 39. Duchen MR. Contributions of mitochondria to animal physiology: from homeostatic sensor to calcium signaling and cell death. J Physiol 516: 1–17, 1999. 40. Dugan LL, Bruno VM, Amagasu SM, and Giffard RG. Glia modulate the response of murine cortical neurons to

URESHINO ET AL.

41.

42.

43. 44.

45.

46.

47.

48. 49. 50.

51. 52.

53.

54. 55. 56.

excitotoxicity: glia exacerbate AMPA neurotoxicity. J Neurosci 15: 4545–4555, 1995. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Khnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, and Shaw RJ. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331: 456–461, 2011. Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, and Nagata S. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391: 43–50, 1998. Fabrizio P, Gattazzo C, Battistella L, Wei M, Cheng C, McGrew K, and Longo VD. Sir2 blocks extreme life-span extension. Cell 123: 655–667, 2005. Feng Z, Hu W, de Stanchina E, Teresky AK, Jin S, Lowe S, and Levine AJ. The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1–AKT–mTOR pathways. Cancer Res 67: 3043–3053, 2007. Ferber EC, Peck B, Delpuech O, Bell GP, East P, and Schulze A. FOXO3a regulates reactive oxygen metabolism by inhibiting mitochondrial gene expression. Cell Death Differ 19: 968–979, 2012. Ferrari D, Pinton P, Szabadadkai G, Chami M, Campanella M, Pozzan T, and Rizzuto R. Endoplasmic reticulum, Bcl-2 and Ca2 + handling in apoptosis. Cell Calcium 32: 413–420, 2002. Filimonenko M, Stuffers S, Raiborg C, Yamamoto A, Malerod L, Fisher EM, Isaacs A, Brech A, Stenmark H, and Simonsen A. Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J Cell Biol 179: 485–500, 2007. Finch CE. Neurons, glia, and plasticity in normal brain aging. Neurobiol Aging Suppl 1: S123–S127, 2003. Finkel T and Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature 408: 239–247, 2000. Fogal B, Trettel J, Uliasz TF, Levine ES, and Hewett SJ. Changes in secondary glutamate release underlie the developmental regulation of excitotoxic neuronal cell death. Neuroscience 132: 929–942, 2005. Foltynie T, Sawcer S, Brayne C, and Barker RA. The genetic basis of Parkinson’s disease. J Neurol Neurosurg Psychiatry 73: 363–370, 2002. Fujiwara M, Marusawa H, Wang HQ, Iwai A, Ikeuchi K, Imai Y, Kataoka A, Nukina N, Takahashi R, and Chiba T. Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene 27: 6002–6011, 2008. Garcı´a-Cao I, Garcı´a-Cao M, Martı´n-Caballero J, Criado LM, Klatt P, Flores JM, Weill JC, Blasco MA, and Serrano M. ‘‘Super p53’’ mice exhibit enhanced DNA damage response, are tumor resistant and age normally. EMBO J 21: 6225–6235, 2002. Gershanik OS. Early onset parkinsonism. Front Biosci 8: 568–578, 2003. Gomes LC and Scorrano L. Mitochondrial elongation during autophagy: a stereotypical response to survive in difficult times. Autophagy 7: 1251–1253, 2011. Granville DJ, Ruehlmann DO, Choy JC, Cassidy BA, Hunt DWC, van Breemen C, and McManus BM. Bcl-2 increases emptying of endoplasmic reticulum Ca2 + stores

CALCIUM, ROS, AUTOPHAGY, AND AGING

57. 58.

59. 60.

61.

62. 63. 64.

65. 66. 67.

68.

69.

70.

71.

72.

during photodynamic therapy-induced apoptosis. Cell Calcium 30: 343–350, 2001. Green DR and Kroemer G. Cytoplasmic functions of the tumour suppressor p53. Nature 458: 1127–1130, 2009. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, MartinMontalvo A, Saavedra J, Ingles S, de Cabo R, Cohen P, and Longo VD. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med 3: 70ra13, 2011. Gunter TE, Buntina L, Sparagna G, Eliseev R, and Gunter K. Mitochondrial calcium transport: mechanisms and functions. Cell Calcium 28: 285–296, 2000. Hagen TM, Yowe DL, Bartholomew JC, Wehr CM, Do KL, Park JY, and Ames BN. Mitochondrial decay in hepatocytes from old rats: membrane potential declines, heterogeneity and oxidants increase. Proc Natl Acad Sci U S A 94: 3064–3069, 1997. Hagenbuchner J, Kuznetsov A, Hermann M, Hausott B, Obexer P, and Ausserlechner MJ. FOXO3-induced reactive oxygen species are regulated by BCL2L11 (Bim) and SESN3. J Cell Sci 125: 1191–1203, 2012. Haigis M and Guarente LP. Mammalian sirtuins-emerging roles in physiology, aging, and calorie restriction. Genes Develop 20: 2913–2921, 2006. Haigis MC and Sinclair DA. Mammalian sirtuins: biological insights and disease relevance. Ann Rev Pathol 5: 253–295, 2010. Hansson MJ, Ma˚nsson R, Morota S, Uchino H, Kallur T, Sumi T, Ishii N, Shimazu M, Keep MF, Jegorov A, and Elme´r E. Calcium-induced generation of reactive oxygen species in brain mitochondria is mediated by permeability transition. Free Radic Biol Med 45: 284–294, 2008. Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol 11: 298–300, 1956. Harman D. The biologic clock: the mitochondria? J Am Geriatr Soc 20: 145–147, 1972. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, and Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460: 392–395, 2009. Herrington J, Park YB, Babcock DF, and Hille B. Dominant role of mitochondria in clearance of large Ca2 + loads from rat adrenal chromaffin cells. Neuron 16: 219–228, 1996. Hildeman DA, Mitchell T, Aronow B, Wojciechowski S, Kappler J, and Marrack P. Control of Bcl-2 expression by reactive oxygen species. Proc Natl Acad Sci U S A 100: 15035–15040, 2003. Hirata H, Lopes GS, Jurkiewicz A, Garcez-do-Carmo L, and Smaili SS. Bcl-2 modulates endoplasmic reticulum and mitochondrial calcium stores in PC12 cells. Neurochem Res 37: 238–243, 2012. Hou X, Xu S, Maitland-Toolan M, Sato K, Jiang B, Ido Y, Lan F, Walsh K, Wierzbicki M, Verbeuren TJ, Cohen RA, and Zang M. SIRT 1 regulates hepatocyte lipid metabolism through activing AMP-actived protein kinase. J Biol Chem 283: 20015–20026, 2008. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, and Sinclair DA. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425: 191–196, 2003.

133

73. Hsu Y, Wolter KG, and Youle RJ. Cytosol-to-membrane redistribution of Bax and Bcl-xL during apoptosis. Proc Natl Acad Sci U S A 94: 3668–3672, 1997. 74. Humpert PM, Djuric Z, Zeuge U, Oikonomou D, Seregin Y, Laine K, Eckstein V, Nawroth PP, and Bierhaus A. Insulin stimulates the clonogenic potential of angiogenic endothelial progenitor cells by IGF-1 receptor-dependent signaling. Mol Med 14: 301–308, 2008. 75. Hussain SP, Amstad P, He P, Robles A, Lupold S, Kaneko I, Ichimiya M, Sengupta S, Mechanic L, Okamura S, Hofseth LJ, Moake M, Nagashima M, Forrester KS, and Harris CC. p53-induced up-regulation of MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer Res 64: 2350–2356, 2004. 76. Ibarra C, Estrada M, Carrasco L, Chiong M, Liberona JL, Cadenas C, Dı´az-Araya G, Jaimovich E, and Lavandero S. Insulin-like growth fator-1 induces an inositol 1,4,5risphosphate-dependent increase in nuclear and cytosolic calcium in cultured rat cardiac myocytes. J Biol Chem 279: 7554–7565, 2004. 77. Jia K and Levine B. Autophagy is required for dietary restriction-mediated life span extension in C. elegans. Autophagy 3: 597–599, 2007. 78. Jorda´n J, Galindo NF, Gonza´lez-Garcia C, and Cen˜a V. Role and regulation of p53 in depolarization-induced neuronal death. Neuroscience 122: 707–715, 2003. 79. Kannurpatti SS, Joshi PG, and Joshi NB. Calcium sequestering ability of mitochondria modulates influx of calcium through glutamate receptor channel. Neurochem Res 25: 1527–1536, 2000. 80. Kasapoglu M and Ozben T. Alterations of antioxidant enzymes and oxidative stress markers in aging. Exp Gerontol 36: 209–220, 2001. 81. Kenyon C, Chang J, Gensch E, Rudner A, and Tabtiang R. A C. elegans mutant that lives twice as long as wild type. Nature 366: 461–464, 1993. 82. Kerr JF, Wyllie AH, and Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26: 239–257, 1972. 83. Khachaturian ZS. The role of calcium regulation in brain aging: reexamination of a hypothesis. Aging (Milano) 1: 17–34, 1989. 84. Khan AS, Sane DC, Wannenburg T, and Sonntag WE. Growth hormone, insulin-like growth factor-1 and the aging cardiovascular system. Cardiovasc Res 54: 25–35, 2002. 85. Kim KS, Seu YB, Baek S, Kim MJ, Kim KJ, Kim JH, and Kim JR. Induction of cellular senescence by insulin-like growth factor binding protein-5 through a p53-dependent mechanism. Mol Biol Cell 18: 4553–4552, 2007. 86. Kirischuk S and Verkhratsky A. Calcium homeostasis in aged neurones. Life Sci 59: 451–459, 1996. 87. Kirkwood TB. DNA, mutations and aging. Mutat Res 219: 1–7, 1989. 88. Klionsky DJ. Autophagy: from phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol 8: 931–937, 2007. 89. Kops GJ, Dansen TB, Polderman PE, Saarloos I, Wirtz KW, Coffer PJ, Huang TT, Bos JL, Medema RH, and Burgering BM. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419: 316–321, 2002. 90. Kowaltowski AJ, Cosso RG, Campos CB, and Fiskum G. Effect of Bcl-2 overexpression on mitochondrial structure and function. J Biol Chem 277: 42802–42807, 2002.

134

91. Kurz T, Terman A, Gustafsson B, and Bru¨nk UT. Lysosomes and oxidative stress in aging and apoptosis. Biochim Biophys Acta 1780: 1291–1303, 2008. 92. Lan F, Cacicedo JM, Ruderman N, and Ido Y. SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMPactivated protein kinase activation. J Biol Chem 283: 27628–27635, 2008. 93. Latonen L, Taya Y, and Laiho M. UV-radiation induces dose-dependent regulation of p53 response and modulates p53-HDM2 interaction in human fibroblasts. Oncogene 20: 6784–6793, 2001. 94. Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos M, Alt FW, and Finkel T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci U S A 105: 3374–3379, 2008. 95. Lee S, Jeong SY, Lim WC, Kim S, Park YY, Sun X, Youle RJ, and Cho H. Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate cellular senescence. J Biol Chem 282: 22977–22983, 2007. 96. LeRoith D, Adamo M, Werner H, and Roberts CT, Jr. IGFs and their receptors as growth regulators in physiology and pathological states. Trends Endocrinol Metab 2: 134–139, 1991. 97. Li Q, Wu S, Li SY, Lopez FL, Du M, Kajstura J, Anversa P, and Ren J. Cardiac-specific overexpression of insulin like growth factor 1 attenuates aging- associated cardiac diastolic contractile dysfunction and protein damage. Am J Physiol 292: H1398–H1403, 2007. 98. Li Y, Corradetti MN, Inoki K, and Guan KL. TSC2: filling the GAP in the mTOR signaling pathway. Trends Biochem Sci 29: 32–38, 2004. 99. Li Y, Xu W, McBurney MW, and Longo VD. SirT1 inhibition reduces IGF-I/IRS-2/Ras/ERK1/2 signaling and protects neurons. Cell Metab 8: 38–48, 2008. 100. Lin J. Yang Q, Wilder PT, Carrier F, and Weber DJ. The calcium-binding protein S100B down-regulates p53 and apoptosis in malignant melanoma. J Biol Chem 285: 27487– 27498, 2010. 101. Ling D, Song H, Garza D, Neufeld TP, and Salvaterra PM. Abeta42-induced neurodegeneration via an age-dependent autophagic-lysosomal injury in Drosophila. PloS One 4: e4201, 2009. 102. Linnane AW, Marzuki S, Ozawa T, and Tanaka M. Mitochondrial DNA mutations as an important contributor to ageing and degenerative diseases. Lancet 1: 642–645, 1989. 103. Longo VD. Linking sirtuins, IGF-I signaling, and starvation. Exp Gerontol 44: 70–74, 2009. 104. Lopes GS, Mora OA, Cerri P, Faria FP, Jurkiewicz NH, Jurkiewicz A, and Smaili SS. Mitochondrial alterations and apoptosis in smooth muscle from aged rats. Biochim Biophys Acta 1658: 187–194, 2004. 105. Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, and Yankner BA. Gene regulation and DNA damage in the ageing human brain. Nature 429: 883–891, 2004. 106. Macip S, Igarashi M, Berggren P, Yu J, Lee SW, and Aaronson SA. Influence of induced reactive oxygen species in p53-mediated cell fate decisions. Mol Cell Biol 23: 8576–8585, 2003. 107. Madan E, Gogna R, Bhatt M, Uttam P, Kuppusamy P, and Mahdi AA. Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget 2: 948–957, 2011.

URESHINO ET AL.

108. Maiuri MC, Galluzzi L, Morselli E, Kepp O, Malik SA, and Kroemer G. Autophagy regulation by p53. Curr Opin Cell Biol 22: 181–185, 2010. 109. Mao L, Zabel C, Wacker MA, Nebrich G, Sagi D, Schrade P, Bachmann S, Kowald A, and Klose J. Estimation of the mtDNA mutation rate in aging mice by proteome analysis and mathematical modeling. Exp Gerontol 41: 11–24, 2006. 110. Marin˜o G, Madeo F, and Kroemer G. Autophagy for tissue homeostasis and neuroprotection. Curr Opin Cell Biol 23: 198–206, 2011. 111. Masoro EJ. Caloric restriction-induced life extension of rats and mice: a critique of proposed mechanisms. Biochim Biophys Acta 1790: 1040–1048, 2009. 112. Matheu A, Maraver A, Klatt P, Flores I, Garcia-Cao I, Borras C, Flores JM, Vin˜a J, Blasco MA, and Serrano M. Delayed ageing through damage protection by the Arf/p53 pathway. Nature 448: 375–379, 2007. 113. Matoba S, Kang J, Patino WD, Wragg A, Boehm M, Gavrilova O, Hurley PJ, Bunz F, and Hwang PM. p53 regulates mitochondrial respiration. Science 312: 1650– 1653, 2006. 114. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Sou YS, Saiki S, Kawajiri S, Sato F, Kimura M, Komatsu M, Hattori N, and Tanaka K. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol 189: 211–221, 2010. 115. Mattson MP, Duan W, and Guo Z. Meal size and frequency affect neuronal plasticity and vulnerability to disease: cellular and molecular mechanisms. J. Neurochem 84: 417–431, 2003. 116. McCammon MT, Veenhuis M, Trapp SB, and Goodman JM, Association of glyoxylate and beta-oxidation enzymes with peroxisomes of Saccharomyces cerevisiae. J Bacteriol 172: 5816–5827, 1990. 117. McCay CM, Crowell MF, and Maynard LA. The effect of retarded growth upon the length of life span and upon the ultimate body size. J Nutr 10: 63–79, 1935. 118. Michain S and Sinclair D. Sirtuins in mammals: insights into their biological function. Biochem J 404: 1–13, 2007. 119. Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, and Moll UM. p53 has a direct apoptogenic role at the mitochondria. Mol Cell 11: 577–590, 2003. 120. Miura D, Miura Y, and Yagasaki K. Hypolipidemic action of dietary resveratrol, a phytoalexin in grapes and red wine, in hepatoma-bearing rats. Life Sci 73: 1393–1400, 2003. 121. Miyashita T and Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80: 293–299, 1995. 122. Morgan TE, Rozovsky I, Goldsmith SK, Stone DJ, Yoshida T, and Finch CE. Increased transcription of the astrocyte gene GFAP during middle-age is attenuated by food restriction: implications for the role of oxidative stress. Free Radic Biol Med 23: 524–528, 1997. 123. Morselli E, Maiuri MC, Markaki M, Megalou E, Pasparaki A, Palikaras K, Criollo A, Galluzzi L, Malik AS, Vitale I, Michaud M, Madeo F, Tavernarakis N, and Kroemer G. Caloric restriction and resveratrol promote longevity through the Sirtuin 1-dependent induction of autophagy. Cell Death Dis 1: e10, 2010. 124. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M, and Guarente L. Mammalian

CALCIUM, ROS, AUTOPHAGY, AND AGING

125. 126. 127. 128.

129.

130.

131.

132. 133. 134.

135.

136. 137. 138.

139. 140.

141.

SIRT1 represses forkhead transcription factors. Cell 116: 551–563, 2004. Mullart E, Lohman PHM, Berends F, and Vijg J. DNA damage metabolism and aging. Mutat Res 237: 189–210, 1990. Muller F. The nature and mechanism of superoxide production by the electron transport chain: its relevance to aging. J Am Aging Assoc 23: 227–253, 2000. Murchison D and Griffith WH. Increased calcium buffering in basal forebrain neurons during aging. J Neurophysiol 80: 350–364, 1998. Myers MG Jr, Backer JM, Sun XJ, Shoelson S, Hu P, Schlessinger J, Yoakim M, Schaffhausen B, and White MF. IRS-1 activates phosphatidylinositol 3¢-kinase by associating with src homology 2 domains of p85. Proc Natl Acad Sci U S A 89: 10350–10354, 1992. Myers MG Jr, Grammer TC, Wang LM, Sun XJ, Pierce JH, Blenis J, and White MF. Insulin receptor substrate-1 mediates phosphatidylinositol 3’-kinase and p70S6k signaling during insulin, insulin-like growth factor-1, and interleukin-4 stimulation. J Biol Chem 69: 28783–28789, 1994. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, and Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 8: e1000298, 2010. Navarro A, Lo´pez-Cepero JM, Ba´ndez MJ, Sa´nchez-Pino MJ, Go´mez C, Cadenas E, and Boveris A. Hippocampal mitochondrial dysfunction in rat aging. Am J Physiol Regul Integr Comp Physiol 294: 501–509, 2007. Olney JW. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science 164: 719–721, 1969. Orrenius S, Zhivotovsky B, and Nicotera P. Regulation of cell death: the calcium-apoptosis link. Nat Rev Mol Cell Biol 4: 552–565, 2003. Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, Ke H, Rehmann H, Taussig R, Brown AL, Kim MK, Beaven MA, and Burgin AB. Resveratrol ameliorates aging- related metabolic phenotypes by inhibition cAMP phosphodiesterases. Cell 148: 421–433, 2012. Pawlikowska L, Hu D, Huntsman S, Sung A, Chu C, Chen J, Joyner AH, Schork NJ, Hsueh WC, Reiner AP, Psaty BM, Atzmon G, Barzilai N, Cummings SR, Browner WS, Kwok PY, and Ziv E. Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell 8: 460–472, 2009. Peng TI, Jou MJ, Sheu SS, and Greenamyre JT. Visualization of NMDA receptor-induced mitochondrial calcium accumulation in striatal neurons. Exp Neurol 149: 1–12, 1998. Pe´richon R, Bourre JM, Kelly JF, and Roth GS. The role of peroxisomes in aging. Cell Mol Life Sci 54: 641–652, 1998. Pinton P, Pozzan T, and Rizzuto R. The Golgi apparatus is an inositol 1,4,5-trisphosphate-sensitive Ca2 + store, with functional properties distinct from those of the endoplasmic reticulum. EMBO J 17: 5298–5308, 1998. Polyak K, Xia Y, Zeiweier YL, Kinzler KW, and Volgestein B. A model for p53-induced apoptosis. Nature 389: 300–305, 1997. Rajdev S and Reynolds IJ. Glutamate induced intracellular calcium changes and neurotoxicity in cortical neurons in vitro: effect of chemical ischemia. Neuroscience 62: 667–679, 1994. Renault VM, Rfafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE, Villeda SA, Thekkat PU, Guillerey C,

135

142.

143. 144. 145.

146. 147. 148.

149.

150.

151.

152.

153. 154.

155.

156.

157.

Denko NC, Palmer TD, Butte AJ, and Brunet A. FoxO3 regulates neural stem cell homeostasis. Stem Cell 5: 527– 539, 2009. Resnicoff M, Abraham D, Yutanawiboonchai W, Rotman HL, Kajsura J, Rubin R, Zolticj R, and Baserga R. The insulin-like growth factor-1 receptors protects tumor cells from apoptosis in vivo. Cancer Res 55: 2463–2469, 1995. Rizzuto R and Pozzan T. Microdomains of intracellular Ca2 + : molecular determinants and functional consequences. Physiol Rev 86: 369–408, 2006. Rollo CD, Carlson J, and Sawada M. Accelerated aging of giant transgenic mice is associated with elevated free radicals process. Can J Zool 74: 606–620, 1996. Rong YP, Bultynck G, Aromolaran AS, Zhong F, Parys JB, De Smedt H, Mignery GA, Roderick HL, Bootman MD, and Distelhorst CW. The BH4 domain of Bcl-2 inhibits ER calcium release and apoptosis by binding the regulatory and coupling domain of the IP3 receptor. Proc Natl Acad Sci U S A 106: 14397–14402, 2009. Rubinsztein DC, Marin˜o G, and Kroemer G. Autophagy and aging. Cell 2: 682–695, 2011. Ruderman NB, Kapeller R, White MF, and Cantley LC. Activation of phosphatidylinositol 3-kinase by insulin. Proc Natl Acad Sci U S A 87:1411–1415, 1990. Sablina AA, Budanov AV, Ilyinskaya GV, Agapova LS, Kravchenko JE, and Chumakov PM. The antioxidant function of the p53 tumor suppressor. Nat Med 11: 1306– 1313, 2005. Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond M, Coskun PE, Ladiges W, Wolf N, Van Remmen H, Wallace DC, and Rabinovitch PS. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science 308: 1909–1911, 2005. Seo AY, Joseph AM, Dutta D, Hwang JC, Aris JP, and Leeuwenburgh C. New insights into the role of mitochondria in aging: mitochondrial dynamics and more. J Cell Sci 123: 2533–2542, 2010. Shan H, Messi ML, Zheng Z, Wang ZM, and Delbono O. Preservation of motor neuron Ca2 + channel sensitivity to insulin-like growth factor-1 in brain motor cortex from senescent rat. J Physiol 553: 49–63, 2003. Sharp ZD and Bartke A. Evidence for downregulation of phosphoinositide 3- kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR)-dependent translation regulatory signaling pathways in Ames dwarf mice. J Gerontol A Biol Sci Med Sci 60: 293–300, 2005. Sinclair DA. Toward a unified theory of caloric restriction and longevity regulation. Mech Ageing Dev 126: 987– 1002, 2005. Siqueira IR, Fochesatto C, de Andrade A, Santos M, Hagen M, Bello-Klein A, and Netto CA. Total antioxidant capacity is impaired in different structures from aged rat brain. Int J Dev Neurosci 23: 663–671, 2005. Smaili SS, Hsu YT, Carvalho AC, Rosenstock TR, Sharpe JC, and Youle RJ. Mitochondria, calcium and pro-apoptotic proteins as mediators in cell death signaling. Braz J Med Biol Res 36: 183–190, 2003. Smaili SS and Russell JT. Permeability transition pore regulates both mitochondrial membrane potential and agonist-evoked Ca2 + signals in oligodendrocyte progenitors. Cell Calcium 26: 121–130, 1999. Smaili SS, Ureshino RP, Rodrigues L, Rocha KK, Carvalho JT, Oseki KT, Bincoletto C, Lopes GS, and Hirata H. The role of mitochondrial function in glutamate-

136

158. 159. 160.

161.

162.

163.

164. 165.

166. 167.

168.

169.

170.

171.

URESHINO ET AL.

dependent metabolism in neuronal cells. Curr Pharm Des 17: 3865–3877, 2011. Sohal RS and Weindruch R. Oxidative stress, caloric restriction and aging. Science 273: 59–63, 1996. Sonntag WE, Ramsey M, and Carter CS. Growth hormone and insulin like growth factor-1 (IGF-1) and their influence on cognitive aging. Ageing Res Rev 4: 195–212, 2005. Suh Y, Atzmon G, Cho MO, Hwang D, Liu B, Leahy DJ, Barzilai N, and Cohen P. Functionally significant insulinlike growth factor I receptor mutations in centenarians. Biol Sci Gen 105: 3438–3442, 2008. Sulaiman M, Matta MJ, Sunderesan NR, Gupta MP, Periasamy M, and Gupta M. Resveratrol, an activator of SIRT1, upregulates sarcoplasmic calcium ATPase and improves cardiac function in diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 298: H833–H843, 2010. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loefer M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, and Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397: 441–446, 1999. Tan M, Li S, Swaroop M, Guan K, Oberley LW, and Sun Y. Transcriptional activation of the human glutathione peroxidase promoter by p53. J Biol Chem 274: 12061– 12066, 1999. Tan WQ, Wang K, Lv DY, and Li PF. Foxo3a inhibits cardiomyocyte hypertrophy through transactivating catalase. J Biol Chem 283: 29730–29739, 2008. Tasdemir E, Maiuri MC, Galluzzi L, Vitale I, DjavaheriMergny M, D’Amelio M, Criollo A, Morselli E, Zhu C, Harper F, Nannmark U, Samara C, Pinton P, Vicencio JM, Carnuccio R, Moll UM, Madeo F, Paterlini-Brechot P, Rizzuto R, Szabadkai G, Pierron G, Blomgren K, Tavernarakis N, Codogno P, Cecconi F, and Kroemer G. Regulation of autophagy by cytoplasmic p53. Nat Cell Biol 10: 676–687, 2008. Terlecky SR, Koepke JI, and Walton PA. Peroxisomes and aging. Biochim Biophys Acta 1763: 1749–1754, 2006. Territo PR, Mootha VK, French SA, and Balaban RS. Ca(2 + ) activation of heart mitochondrial oxidative phosphorylation: role of the F(0)/F(1)-ATPase. Am J Physiol Cell Physiol 278: 423–435, 2000. Thees S, Hubbard GB, Winckler J, Schultz C, and Rami A. Specific alteration of the Bax/Bcl2 ratio and cytochrome c without execution of apoptosis in the hippocampus of aged baboons. Restor Neurol Neurosci 23: 1–9, 2005. Tian L, Cai Q, and Wei H. Alterations of antioxidant enzymes and oxidative damage to macromolecules in different organs of rats during aging. Free Radic Biol Med 24: 1477–1484, 1998. Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, Hoeks J, van der Krieken S, Ryu D, Kersten S, Moonen-Komips E, Hesselink MKC, Kunz I, Schrauwen-Hinderling VB, Blaak EE, Auwerx J, and Schrauwen P. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab 14: 612– 622, 2011. Tong C, Morrison A, Mattison S, Qian S, Bryniarski M, Rankin B, Wang J, Thomas P, and Li J. Impaired SIRT 1 nucleocytoplasm shuttling in the senescent heart during ischemic stress. FASEB J 27: 1–11, 2013.

172. Torella D, Rota M, Nurzynska D, Musso E, Monsen A, Shiraishi I, Zias E, Walsh K, Rosenzweig A, Sussman MA, Urbanek K, Nadal-Ginard B, Kajstura J, Aversa P, and Leri A. Cardiac stem cell and myocyte aging heart failure and insulin-like growth factor-1 overexpression. Circ Res 94: 514–524, 2004. 173. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, Bruder CE, Bohlooly MY, Gidlof S, Oldfors A, Wibom R, Tornell J, Jocobs HT, and Larssom N. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429: 417–423, 2004. 174. Tyner SD, Venkatachalam S, Choi J, Jones S, Ghebranious N, Igelmann H, Lu X, Soron G, Cooper B, Brayton C, Park SH, Thompson T, Karsenty G, Bradley A, and Donehower LA. p53 mutant mice that display early ageing-associated phenotypes. Nature 415: 45–53, 2002. 175. Ungvari Z, Kaley G, Cabo R, Sonntag WE, and Csiszar A. Mechanisms of vascular: aging: new perspectives. J Gerontol A Biol Sci Med Sci 65: 1028–1041, 2010. 176. Ungvari Z, Orosz Z, Rivera A, Labinskyy N, Xiangmin Z, Olson S, Podlutsky A, and Csiszar A. Resveratrol increases vascular oxidative stress resistance. Am J Physiol Heart Circ Physiol 292: H2417–H2424, 2007. 177. Ureshino RP, Bertoncini CR, Fernandes MJ, Abdalla FM, Porto CS, Hsu YT, Lopes GS, and Smaili SS. Alterations in calcium signaling and a decrease in Bcl-2 expression: possible correlation with apoptosis in aged striatum. J Neurosci Res 88: 438–447, 2010. 178. Veenhuis M, Mateblowski M, Kunau WH, and Harder W. Proliferation of microbodies in Saccharomyces cerevisiae. Yeast 3: 77–84, 1987. 179. Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, Janakiraman M, Schultz N, Hanrahan AJ, Pao W, Ladanyi M, Sander C, Heguy A, Holland EC, Paty PB, Mischel PS, Liau L, Cloughesy TF, Mellinghoff IK, Solit DB, and Chan TA. Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet 42: 77–82, 2010. 180. Vellai T. Autophagy and ageing. Cell Death Differ 16: 94–102, 2009. 181. Vercesi AE, Kowaltowski AJ, Grijalba MT, Meinicke AR, and Castilho RE. The role of reactive oxigen species in mitochondrial permeability transition. Biosci Rep 17: 43– 52, 1997. 182. Verkhratsky A, Rodrı´guez JJ, and Parpura V. Neurotransmitters and integration in neuronal-astroglial networks. Neurochem Res 37: 2326–2338, 2012. 183. Vicencio JM, Galluzzi L, Tajeddine N, Ortiz C, Criollo A, Tasdemir E, Morselli E, Ben Younes A, Maiuri MC, Lavandero S, and Kroemer G. Senescence, apoptosis or autophagy? When a damaged cell must decide its path—a mini-review. Gerontology 54: 92–99, 2008. 184. Vijg J, Busuttil RA, Bahar R, and Dolle´ ME. Aging and genome maintenance. Ann NY Acad Sci 1055: 35–47, 2005. 185. Wang C, Chen L, Hou X, Li Z, Kabra N, Ma Y, Nemoto S, Finkel T, Gu W, Cress WD, and Chen J. Interactions between E2F1 and SirT1 regulate apoptotic response to DNA damage. Nat Cell Biol 8: 1025–1031, 2006. 186. Wang ZM, Messi ML, Renganathan M, and Delbono O. Insulin-like growth factor-1 enhances rat skeletal muscle charge movement and L-type Ca2 + channel gene expression. J Physiol 516: 331–341, 1999. 187. Wei H, Wei W, Bredesen DE, and Perry DC. Bcl-2 protects against apoptosis in neuronal cell line caused by

CALCIUM, ROS, AUTOPHAGY, AND AGING

188. 189.

190.

191. 192.

193. 194. 195. 196.

197. 198.

199.

thapsigargin-induced depletion of intracellular calcium stores. J Neurochem 70: 2305–2314, 1998. Wesierska-Gadek J. mTOR and its link to the picture of Dorian Gray-reactivation of mTOR promotes aging. Aging 2: 892–893, 2010. Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, Yano K, Masaki KH, Willcox DC, Rodriguez B, and Curb D. FOXO3a genotype is strongly associated with human longevity. Proc Natl Acad Sci U S A 105: 13987–13992, 2008. Xiong J, Verkhratsky A, and Toescu EC. Changes in mitochondrial status associated with altered Ca2 + homeostasis in aged cerebellar granule neurons in brain slices. J Neurosci 22: 10761–10771, 2002. Yaffe MP. The machinery of mitochondrial inheritance and behavior. Science 283: 1493–1497, 1999. You H, Pellegrini M, Tsuchihara K, Yamamoto K, Hacker G, Erlacher M, Villunger A, and Mak TW. FOXO3adependent regulation of Puma in response to cytokine/ growth factor withdrawal. J Exp Med 203: 1657–1663, 2006. You H, Yamamoto K, and Mak TW. Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a. Proc Natl Acad Sci U S A 103: 9051–9056, 2006. Youle RJ and Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 12: 9–14, 2011. Yuan Z, Zhang X, Sengupta N, Lane WS, and Seto E. SIRT1 regulates the function of the Nijmegen breakage syndrome protein. Mol Cell 27: 149–162, 2007. Zha H, Fisk HA, Yaffe MP, Mahajan N, Herman B, and Reed JC. Structure–function comparisons of the proapoptotic protein Bax in yeast and mammalian cells. Mol Cell Biol 16: 6494–6508, 1996. Zhang J. Resveratrol inhibits insulin responses in a SirT1independent pathway. Biochem J 397: 519–527, 2006. Zheng WH, Kar S, and Quirion R. Stimulation of protein kinase C modulates insulin-like growth factor-1-induced akt activation in PC12 cells. J Biol Chem 275: 13377– 13385, 2000. Zhu L, Ling S, Yu X-D, Venkastesh LK, Subramanian T, Chinnadurai G, and Kuo TH. Modulation of mitochondrial Ca2 + homeostasis by Bcl-2. J Biol Chem 274: 33267– 33273, 1999.

Address correspondence to: Prof. Soraya Soubhi Smaili Department of Pharmacology Federal University of Sa˜o Paulo Rua Treˆs de Maio, n.100 Sa˜o Paulo/SP 04044-020 Brazil E-mail: [email protected]

137

Date of first submission to ARS Central, December 11, 2013; date of final revised submission, January 24, 2014; date of acceptance, February 8, 2014. Abbreviations Used Dwm ¼ mitochondrial membrane potential AMPK ¼ adenosine monophosphate-activated protein kinase ATPs ¼ ATP synthase CAD ¼ caspase-activated DNase CaMKKb ¼ Ca2+ /calmodulin-dependent protein kinase kinase b CNS ¼ central nervous system CR ¼ caloric restriction DDR ¼ DNA damage response ER ¼ endoplasmic reticulum ETC ¼ electron transport chain FCCP ¼ carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone FoxO3 ¼ forkhead box protein O3 GH ¼ growth hormone GPx ¼ glutathione peroxidase iCAD ¼ inhibitor of caspase-activated DNase IGF ¼ insulin-like growth factor IP3 ¼ inositol trisphosphate IP3 R ¼ inositol trisphosphate receptor IRS-1 ¼ insulin receptor substrate 1 LKB1 ¼ liver kinase B1 MnSOD ¼ manganese superoxide dismutase mtDNA ¼ mitochondrial DNA mTOR ¼ mammalian target of rapamycin NAD ¼ nicotinamide adenine dinucleotide NMDA ¼ N-methyl-D-aspartic acid p.p53 ¼ phosphorilated p53 PI3K ¼ phosphoinositide 3-kinase PINK1 ¼ Pten-induced novel kinase 1 PTP ¼ permeability transition pore Rheb ¼ Ras homologue enriched in brain ROS ¼ reactive oxygen species RyR ¼ ryanodine receptors SCO2 ¼ synthesis of cytochrome c oxidase 2 SERCA ¼ sarco-endoplasmic reticulum Ca2+ -ATPase Sesn ¼ sestrin SOD ¼ superoxide dismutase Tap ¼ Tapsigargin TCA ¼ tricarboxylic acid cycle TIGAR ¼ TP53-induced glycolysis regulator TSC2 ¼ tuberous sclerosis protein 2 VOCC ¼ voltage-operated Ca2+ channels

Calcium signaling alterations, oxidative stress, and autophagy in aging.

Aging is a multi-factorial process that may be associated with several functional and structural deficits which can evolve into degenerative diseases...
957KB Sizes 2 Downloads 0 Views